51
|
Interactions of Tofacitinib and Dexamethasone on Lymphocyte Proliferation. Pharm Res 2020; 37:105. [DOI: 10.1007/s11095-020-02827-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/17/2020] [Indexed: 12/31/2022]
|
52
|
Quatrini L, Vacca P, Tumino N, Besi F, Di Pace AL, Scordamaglia F, Martini S, Munari E, Mingari MC, Ugolini S, Moretta L. Glucocorticoids and the cytokines IL-12, IL-15, and IL-18 present in the tumor microenvironment induce PD-1 expression on human natural killer cells. J Allergy Clin Immunol 2020; 147:349-360. [PMID: 32417134 DOI: 10.1016/j.jaci.2020.04.044] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/28/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Programmed cell death protein 1 (PD-1)-immune checkpoint blockade has provided significant clinical efficacy across various types of cancer by unleashing both T and natural killer (NK) cell-mediated antitumor responses. However, resistance to immunotherapy occurs for many patients, rendering the identification of the mechanisms that control PD-1 expression extremely important to increase the response to the therapy. OBJECTIVE We sought to identify the stimuli and the molecular mechanisms that induce the de novo PD-1 expression on human NK cells in the tumor setting. METHODS NK cells freshly isolated from peripheral blood of healthy donors were stimulated with different combinations of molecules, and PD-1 expression was studied at the mRNA and protein levels. Moreover, ex vivo analysis of tumor microenvironment and NK cell phenotype was performed. RESULTS Glucocorticoids are indispensable for PD-1 induction on human NK cells, in cooperation with a combination of cytokines that are abundant at the tumor site. Mechanistically, glucocorticoids together with IL-12, IL-15, and IL-18 not only upregulate PDCD1 transcription, but also activate a previously unrecognized transcriptional program leading to enhanced mRNA translation and resulting in an increased PD-1 amount in NK cells. CONCLUSIONS These results provide evidence of a novel immune suppressive mechanism of glucocorticoids involving the transcriptional and translational control of an important immune checkpoint.
Collapse
Affiliation(s)
- Linda Quatrini
- Department of Immunology, Istituto di Ricovero e Cura a Carattere Scientifico Bambino Gesù Children's Hospital, Rome, Italy.
| | - Paola Vacca
- Department of Immunology, Istituto di Ricovero e Cura a Carattere Scientifico Bambino Gesù Children's Hospital, Rome, Italy
| | - Nicola Tumino
- Department of Immunology, Istituto di Ricovero e Cura a Carattere Scientifico Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesca Besi
- Department of Immunology, Istituto di Ricovero e Cura a Carattere Scientifico Bambino Gesù Children's Hospital, Rome, Italy
| | - Anna Laura Di Pace
- Department of Immunology, Istituto di Ricovero e Cura a Carattere Scientifico Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Stefania Martini
- Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genoa, Italy
| | - Enrico Munari
- Department of Pathology, Sacro Cuore Don Calabria, Negrar, Italy
| | - Maria Cristina Mingari
- Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genoa, Italy; Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Sophie Ugolini
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Istitut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Lorenzo Moretta
- Department of Immunology, Istituto di Ricovero e Cura a Carattere Scientifico Bambino Gesù Children's Hospital, Rome, Italy.
| |
Collapse
|
53
|
A Strategy for Personalized Treatment of iPS-Retinal Immune Rejections Assessed in Cynomolgus Monkey Models. Int J Mol Sci 2020; 21:ijms21093077. [PMID: 32349277 PMCID: PMC7247695 DOI: 10.3390/ijms21093077] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/25/2020] [Accepted: 04/26/2020] [Indexed: 11/17/2022] Open
Abstract
Recently, we successfully transplanted an autograft, or major histocompatibility complex (MHC)-matched allografts, from induced-pluripotent-stem-cell-derived retinal pigment epithelial (iPSC-RPE) cells in patients with age-related macular degeneration. However, there was an issue regarding immune rejection after transplantation. In this study, we established a preoperational in vitro "drug-lymphocytes-grafts immune reaction (Drug-LGIR)" test to determine the medication for immune rejection using host immunocompetent cells (lymphocytes) and transplant cells (target iPSC-RPE cells) together with different medications. The adequacy of the test was assessed by in vivo transplantation in monkey models together with medication based on in vitro data. In the results of Drug-LGIR tests, some drugs exhibited significant suppression of RPE cell-related allogeneic reactions, while other drugs did not, and the efficacy of each drug differed among the recipient monkeys. Based on the results of Drug-LGIR, we applied cyclosporine A or local steroid (triamcinolone) therapy to two monkeys, and successfully suppressed RPE-related immune rejections with RPE grafts, which survived without any signs of rejection under drug administration. We propose that our new preoperational in vitro Drug-LGIR test, which specifies the most efficacious medication for each recipient, is useful for controlling immune attacks with personalized treatment for each patient after retinal transplantation.
Collapse
|
54
|
The glucocorticoids prednisone and dexamethasone differentially modulate T cell function in response to anti-PD-1 and anti-CTLA-4 immune checkpoint blockade. Cancer Immunol Immunother 2020; 69:1423-1436. [PMID: 32246174 DOI: 10.1007/s00262-020-02555-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/26/2020] [Indexed: 12/14/2022]
Abstract
On-treatment steroids for countering immune checkpoint inhibitor-induced inflammatory responses (irAEs) are a hallmark of cancer immunotherapy. However, the suppressive nature of steroids has raised questions regarding their ability to compromise the function of the 'proliferative burst' of effector T cells induced by immune checkpoint antibodies. We investigated the effector functions and the co-inhibitory receptor profile of stimulated peripheral blood mononuclear cells (PBMCs) pre-treated with prednisone and dexamethasone alone or in the presence of anti-PD-1/CTLA-4 antibodies. Also, clinical analysis of a patient who exhibited irAEs following combination (anti-PD-1/CTLA-4) in the presence of glucocorticoids was done. We found that prednisone in contrast to dexamethasone did not compromise T cell cytokine production (IL-2, IFN-γ and TNF-α) and proliferation in the absence or presence of anti-PD-1/CTLA-4 antibodies, when a physiological concentration was used. Neither single prednisone treatment nor co-treatment with checkpoint inhibitors impacted the expression of co-inhibitory receptors PD-1, CTLA-4, TIM-3 and LAG-3. In contrast, dexamethasone treatment promoted downregulation of LAG-3 expression by T cells. In addition, co-treatment of PD-1 + Jurkat cells with prednisone and/or dexamethasone with anti-PD-1 before stimulation significantly reduced SHP-2 phosphorylation, indicative of increased T cell function. Our findings hereby demonstrate a differential steroid effect on T cell function, which should be taken into consideration for patients undergoing immunotherapy. Also, the clinical analysis of a patient who exhibited irAEs following combination (anti-PD-1/CTLA-4) therapy indicated complete metabolic response in the presence of glucocorticoids. Therefore, concomitant use of prednisone does not appear to interfere with the function of immune checkpoint blockade.
Collapse
|
55
|
Abstract
Given its poor prognosis, glioblastoma represents an area of high unmet clinical need. Standard of care for the treatment of glioblastoma in the frontline setting is limited to surgical resection, radiation, and temozolomide, with the more recent addition of Tumor Treating Fields. Several agents, including bevacizumab, lomustine, and carmustine have been approved in the recurrent setting. To date, no therapies have demonstrated substantial survival benefit beyond standard of care. An expanding understanding of the role of the immune system in fighting cancer has led to the development and approval of various immunotherapeutic approaches across solid tumors. In glioblastoma, the notion of a highly immune-restricted central nervous system has also evolved, further providing the rationale for testing therapies that promote immune trafficking to the CNS and infiltration into the tumor to counteract the immunosuppressive mechanisms that support tumor progression. There are five broad categories of immunotherapies currently being tested in GBM: vaccines, cytokine therapy, oncolytic viral therapy, chimeric antigen receptor T cell therapy, and checkpoint inhibitors. This review focuses on checkpoint inhibitors in GBM, the rationale for its use, preclinical data, and early clinical experience. Efficacy data are limited, and while a number of late-stage trials are ongoing, early trials showed no benefit in survival. There is a dizzying array of combinations being tested in clinical studies with an urgent need for a rational approach to determine the role of checkpoint inhibitors in glioblastoma, including the optimal combinations, and identification of biomarkers or predictive models to determine which patients may benefit from immunotherapy.
Collapse
|
56
|
Kelly WJ, Gilbert MR. Glucocorticoids and immune checkpoint inhibitors in glioblastoma. J Neurooncol 2020; 151:13-20. [PMID: 32108294 DOI: 10.1007/s11060-020-03439-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/14/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE Immunotherapy, activation of the immune system to target tumor cells, represents a paradigm shift in the treatment of cancer. Immune checkpoint therapies, which target immunomodulatory molecules expressed on T-lymphocytes, have demonstrated improved survival in a variety of malignancies. However, benefit in glioblastoma, the most common and devastating malignant brain tumor, remains to be seen. With several recent clinical trials failing to show efficacy of immunotherapy, concerns have been raised regarding the impact of glucocorticoid use in this patient population that may impair the ability for immune checkpoint inhibitors to affect a response. METHODS For this article we examined the mechanism by which immune checkpoint inhibitors activate, and glucocorticoids impair, T-lymphocyte function. RESULTS In this context, we review the clinical data of immune checkpoint inhibitors in glioblastoma as well as the impact glucocorticoids have on immune checkpoint inhibitor efficacy. Finally, we highlight key questions that remain in the field, and the potential benefit of further research for central nervous system tumors. CONCLUSION More information on the extent, character and duration of glucocorticoids on patients treated with PD-(L)1 will better inform both clinical management and novel therapeutic development of immunotherapy in patients with CNS malignancies.
Collapse
Affiliation(s)
- William J Kelly
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Building 82, Room 235, 9030 Old Georgetown Road, Bethesda, MD, 20892, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Building 82, Room 235, 9030 Old Georgetown Road, Bethesda, MD, 20892, USA.
| |
Collapse
|
57
|
Petrelli F, Signorelli D, Ghidini M, Ghidini A, Pizzutilo EG, Ruggieri L, Cabiddu M, Borgonovo K, Dognini G, Brighenti M, De Toma A, Rijavec E, Garassino MC, Grossi F, Tomasello G. Association of Steroids use with Survival in Patients Treated with Immune Checkpoint Inhibitors: A Systematic Review and Meta-Analysis. Cancers (Basel) 2020; 12:E546. [PMID: 32120803 PMCID: PMC7139305 DOI: 10.3390/cancers12030546] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/23/2020] [Accepted: 02/25/2020] [Indexed: 12/21/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) can elicit toxicities by inhibiting negative regulators of adaptive immunity. Sometimes, management of toxicities may require systemic glucocorticoids. We performed a systematic review and meta-analysis of published studies to evaluate the correlation between steroids use, overall survival (OS), and progression-free survival (PFS) in cancer patients treated with ICIs. Publications that compared steroids with non-steroid users in cancer patients treated with ICIs from inception to June 2019 were identified by searching the EMBASE, PubMed, SCOPUS, Web of Science, and Cochrane Library databases. The pooled hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated using a random-effects model. Patients (studies, n = 16; patients, n = 4045) taking steroids were at increased risk of death and progression compared to those not taking steroids (HR = 1.54, 95% CI: 1.24-1.91; p = 0.01 and HR = 1.34, 95% CI: 1.02-1.76; p = 0.03, respectively). The main negative effect on OS was associated with patients taking steroids for supportive care (HR = 2.5, 95% CI 1.41-4.43; p < 0.01) or brain metastases (HR = 1.51, 95% CI 1.22-1.87; p < 0.01). In contrast, steroids used to mitigate adverse events did not negatively affect OS. In conclusion, caution is needed when steroids are used for symptom control. In these patients, a negative impact of steroid use was observed for both OS and PFS.
Collapse
Affiliation(s)
- Fausto Petrelli
- Medical Oncology Unit, ASST Bergamo Ovest, 24047 Treviglio (BG), Italy; (M.C.); (K.B.)
| | - Diego Signorelli
- Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (D.S.); (A.D.T.); (M.C.G.)
| | - Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milano, Italy; (M.G.); (E.R.); (F.G.)
| | - Antonio Ghidini
- Medical Oncology Unit, Casa di Cura Igea, 20126 Milano, Italy;
| | - Elio Gregory Pizzutilo
- Medical Oncology Unit, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy; (E.G.P.); (L.R.); (G.T.)
| | - Lorenzo Ruggieri
- Medical Oncology Unit, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy; (E.G.P.); (L.R.); (G.T.)
| | - Mary Cabiddu
- Medical Oncology Unit, ASST Bergamo Ovest, 24047 Treviglio (BG), Italy; (M.C.); (K.B.)
| | - Karen Borgonovo
- Medical Oncology Unit, ASST Bergamo Ovest, 24047 Treviglio (BG), Italy; (M.C.); (K.B.)
| | | | | | - Alessandro De Toma
- Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (D.S.); (A.D.T.); (M.C.G.)
| | - Erika Rijavec
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milano, Italy; (M.G.); (E.R.); (F.G.)
| | - Marina Chiara Garassino
- Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (D.S.); (A.D.T.); (M.C.G.)
| | - Francesco Grossi
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milano, Italy; (M.G.); (E.R.); (F.G.)
| | - Gianluca Tomasello
- Medical Oncology Unit, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy; (E.G.P.); (L.R.); (G.T.)
| |
Collapse
|
58
|
Dangi A, Yu S, Lee FT, Burnette M, Wang JJ, Kanwar YS, Zhang ZJ, Abecassis M, Thorp EB, Luo X. Murine cytomegalovirus dissemination but not reactivation in donor-positive/recipient-negative allogeneic kidney transplantation can be effectively prevented by transplant immune tolerance. Kidney Int 2020; 98:147-158. [PMID: 32471635 PMCID: PMC7311252 DOI: 10.1016/j.kint.2020.01.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 12/17/2019] [Accepted: 01/24/2020] [Indexed: 12/12/2022]
Abstract
Cytomegalovirus (CMV) reactivation from latently infected donor organs post-transplantation and its dissemination cause significant comorbidities in transplant recipients. Transplant-induced inflammation combined with chronic immunosuppression has been thought to provoke CMV reactivation and dissemination, although sequential events in this process have not been studied. Here, we investigated this process in a high-risk donor CMV-positive to recipient CMV-negative allogeneic murine kidney transplantation model. Recipients were either treated with indefinite immunosuppression or tolerized in a donor-specific manner. Untreated recipients served as controls. Kidney allografts from both immunosuppressed and tolerized recipients showed minimal alloimmunity-mediated graft inflammation and normal function for up to day 60 post-transplantation. However, despite the absence of such inflammation in the immunosuppressed and tolerized groups, CMV reactivation in the donor positive kidney allograft was readily observed. Interestingly, subsequent CMV replication and dissemination to distant organs only occurred in immunosuppressed recipients in which CMV-specific CD8 T cells were functionally impaired; whereas in tolerized recipients, host anti-viral immunity was well-preserved and CMV dissemination was effectively prevented. Thus, our studies uncoupled CMV reactivation from its dissemination, and underscore the potential role of robust transplantation tolerance in preventing CMV diseases following allogeneic kidney transplantation.
Collapse
Affiliation(s)
- Anil Dangi
- Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Shuangjin Yu
- Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Division of Organ Transplantation, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Frances T Lee
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Melanie Burnette
- Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Jiao-Jing Wang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yashpal S Kanwar
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Zheng J Zhang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michael Abecassis
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Edward B Thorp
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Xunrong Luo
- Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Duke Transplant Center, Duke University Medical Center, Durham, North Carolina, USA.
| |
Collapse
|
59
|
Wofford KL, Singh BS, Cullen DK, Spiller KL. Biomaterial-mediated reprogramming of monocytes via microparticle phagocytosis for sustained modulation of macrophage phenotype. Acta Biomater 2020; 101:237-248. [PMID: 31731024 PMCID: PMC6960335 DOI: 10.1016/j.actbio.2019.11.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/14/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022]
Abstract
Monocyte-derived macrophages orchestrate tissue regeneration by homing to sites of injury, phagocytosing pathological debris, and stimulating other cell types to repair the tissue. Accordingly, monocytes have been investigated as a translational and potent source for cell therapy, but their utility has been hampered by their rapid acquisition of a pro-inflammatory phenotype in response to the inflammatory injury microenvironment. To overcome this problem, we designed a cell therapy strategy where monocytes are exogenously reprogrammed by intracellularly loading the cells with biodegradable microparticles containing an anti-inflammatory drug in order to modulate and maintain an anti-inflammatory phenotype over time. To test this concept, poly(lactic-co-glycolic) acid microparticles were loaded with the anti-inflammatory drug dexamethasone (Dex) and administered to primary human monocytes for four hours to facilitate phagocytic uptake. After removal of non-phagocytosed microparticles, microparticle-loaded monocytes differentiated into macrophages and stored the microparticles intracellularly for several weeks in vitro, releasing drug into the extracellular environment over time. Cells loaded with intracellular Dex microparticles showed decreased expression and secretion of inflammatory factors even in the presence of pro-inflammatory stimuli up to 7 days after microparticle uptake compared to untreated cells or cells loaded with blank microparticles, without interfering with phagocytosis of tissue debris. This study represents a new strategy for long-term maintenance of anti-inflammatory macrophage phenotype using a translational monocyte-based cell therapy strategy without the use of genetic modification. Because of the ubiquitous nature of monocyte-derived macrophage involvement in pathology and regeneration, this strategy holds potential as a treatment for a vast number of diseases and disorders. STATEMENT OF SIGNIFICANCE: We report a unique and translational strategy to overcome the challenges associated with monocyte- and macrophage-based cell therapies, in which the cells rapidly take on inflammatory phenotypes when administered to sites of injury. By intracellularly loading monocytes with drug-loaded microparticles prior to administration via phagocytosis, we were able to inhibit inflammation while preserving functional behaviors of human primary macrophages derived from those monocytes up to seven days later. To our knowledge, this study represents the first report of reprogramming macrophages to an anti-inflammatory phenotype without the use of genetic modification.
Collapse
Affiliation(s)
- Kathryn L Wofford
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States; Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, United States; Departments of Neurosurgery & Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Bhavani S Singh
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - D Kacy Cullen
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, United States; Departments of Neurosurgery & Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Kara L Spiller
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
60
|
Matubu A, Hillier SL, Meyn LA, Stoner KA, Mhlanga F, Mbizvo M, Maramba A, Chirenje ZM, Achilles SL. Depot medroxyprogesterone acetate and norethisterone enanthate differentially impact T-cell responses and expression of immunosuppressive markers. Am J Reprod Immunol 2019; 83:e13210. [PMID: 31729087 DOI: 10.1111/aji.13210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/07/2019] [Accepted: 10/22/2019] [Indexed: 11/28/2022] Open
Abstract
PROBLEM Injectable contraceptive use may impact immune cell responsiveness and susceptibility to infection. We measured responsiveness of T-cells from women before and after initiating depot medroxyprogesterone acetate (DMPA) or norethisterone enanthate (Net-En). METHOD OF STUDY Peripheral blood mononuclear cells collected from women aged 18-34 years prior to, at steady state, and nadir concentrations after initiating DMPA (n = 30) or Net-En (n = 36) and from women initiating copper intrauterine device (CU-IUD; n = 32) were stimulated with phorbol myristate acetate and analyzed using flow cytometry. We evaluated percentage change in T-cells expressing programmed cell death-1 (PD-1) and cytotoxic T-lymphocyte associated protein-4 (CTLA-4). RESULTS Compared to baseline, there were decreased numbers of CD4+CTLA4+ (P < .001) and CD8+CTLA4+ (P < .01) T-cells following ex vivo stimulation challenge at steady state DMPA concentrations and no differences at nadir concentrations (P = .781 and P = .463, respectively). In Net-En users, no differences in CD4+CTLA4+ T-cells at steady state (P = .087) and nadir concentrations (P = .217) were observed. DMPA users had fewer CD4+PD-1+ (P < .001) and CD8+PD-1+ (P < .001) T-cells at nadir concentrations. Number of CD4+PD-1+ and CD8+PD-1+ T-cells decreased at steady state concentration (P = .002 and P = .001, respectively) and at nadir concentrations after Net-En initiation (P < .001 and P < .001). In CU-IUD users, there were no changes in number of CD4+CTLA4+ (P = .426) and CD8+CTLA4+ (P = .169) and no changes in CD4+PD-1+ (P = .083) and CD8+PD-1+ (P = .936) compared to baseline. CONCLUSION Activation of T-cells in response to ex vivo stimulation is suppressed at steady state DMPA concentration and resolves at nadir concentration, suggesting DMPA immunosuppressive effects may be transient.
Collapse
Affiliation(s)
- Allen Matubu
- Department of Obstetrics and Gynaecology, University of Zimbabwe College of Health Sciences-Clinical Trials Research Centre, Harare, Zimbabwe
| | - Sharon L Hillier
- Department of Obstetrics, Gynaecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Leslie A Meyn
- Department of Obstetrics, Gynaecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Felix Mhlanga
- Department of Obstetrics and Gynaecology, University of Zimbabwe College of Health Sciences-Clinical Trials Research Centre, Harare, Zimbabwe
| | - Mike Mbizvo
- Department of Obstetrics and Gynaecology, University of Zimbabwe College of Health Sciences-Clinical Trials Research Centre, Harare, Zimbabwe
| | - Aaron Maramba
- Department of Medical Laboratory Sciences, University of Zimbabwe College of Health Sciences, Harare, Zimbabwe
| | - Zvavahera M Chirenje
- Department of Obstetrics and Gynaecology, University of Zimbabwe College of Health Sciences-Clinical Trials Research Centre, Harare, Zimbabwe
| | - Sharon L Achilles
- Department of Obstetrics, Gynaecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Magee-Womens Research Institute, Pittsburgh, PA, USA
| |
Collapse
|
61
|
Maeda N, Maruhashi T, Sugiura D, Shimizu K, Okazaki IM, Okazaki T. Glucocorticoids potentiate the inhibitory capacity of programmed cell death 1 by up-regulating its expression on T cells. J Biol Chem 2019; 294:19896-19906. [PMID: 31723031 DOI: 10.1074/jbc.ra119.010379] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/08/2019] [Indexed: 01/09/2023] Open
Abstract
The inhibitory co-receptor programmed cell death 1 (PD-1, Pdcd1) plays critical roles in the regulation of autoimmunity, anticancer immunity, and immunity against infections. Immunotherapies targeting PD-1 have revolutionized cancer management and instigated various trials of improved cancer immunotherapies. Moreover, extensive trials are underway to potentiate PD-1 function to suppress harmful immune responses. Here we found that both natural and synthetic glucocorticoids (GCs) up-regulate PD-1 on T cells without altering the expression levels of other co-receptors and cell surface molecules. GC-induced up-regulation of PD-1 depended on transactivation of PD-1 transcription mediated through the glucocorticoid receptor. We further found that a GC response element 2525 bp upstream of the transcription start site of Pdcd1 is responsible for GC-mediated transactivation. We also observed that in vivo administration of GCs significantly up-regulates PD-1 expression on tumor-infiltrating T cells. By analyzing T cells differing in PD-1 expression, we directly demonstrated that the amount of PD-1 on the cell surface correlates with its inhibitory effect. Accordingly, GCs potentiated the capacity of PD-1 to inhibit T cell activation, suggesting that this PD-1-mediated inhibition contributes, at least in part, to the anti-inflammatory and immunosuppressive effects of GCs. In light of the critical roles of PD-1 in the regulation of autoimmunity, we expect that the potentiation of PD-1 activity may offer a promising therapeutic strategy for managing inflammatory and autoimmune diseases. Our current findings provide a rationale for strategies seeking to enhance the inhibitory effect of PD-1 by increasing its expression level.
Collapse
Affiliation(s)
- Natsumi Maeda
- Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8503, Japan
| | - Takumi Maruhashi
- Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8503, Japan.,Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Daisuke Sugiura
- Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8503, Japan.,Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Kenji Shimizu
- Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8503, Japan.,Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Il-Mi Okazaki
- Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8503, Japan.,Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Taku Okazaki
- Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8503, Japan .,Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| |
Collapse
|
62
|
Zhao Y, Jia Y, Shi T, Wang W, Shao D, Zheng X, Sun M, He K, Chen L. Depression promotes hepatocellular carcinoma progression through a glucocorticoid-mediated upregulation of PD-1 expression in tumor-infiltrating NK cells. Carcinogenesis 2019; 40:1132-1141. [PMID: 30715244 DOI: 10.1093/carcin/bgz017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/09/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022] Open
Abstract
There is a growing belief that depression was positively associated with the progression of liver cancer. However, the driving molecular events behind the depression in liver cancer are poorly understood and need to be elucidated. Since hyperactivity of the hypothalamic-pituitary-adrenal axis during depression leads to the excessive release of glucocorticoids (GCs), which suppress the activity of natural killer (NK) cells, we hypothesized that high levels of GCs during depression may inhibit function of tumor-infiltrating NK cells during the progress of the liver cancer. Using chronic unpredictable mild stress-induced depressed mice model, we showed that the progression of liver cancer was significantly accelerated in the depressed mice. The high levels of GCs were observed in both depressed mice and depressed patients with liver cancer. Importantly, the expression of programmed death (PD)-1 on NK cells was specifically increased in the tumor microenvironment rather than that in blood or spleen. Coculture studies demonstrated that the expression of PD-1 was significantly increased and cytotoxicity of NK92 cells was remarkably decreased by the dexamethasone treatment through PD-L1-dependent pathway. To the best of our knowledge, we first found that PD-1/PD-L1-mediated exhaustion of infiltrated NK cells promoted hepatocellular carcinoma progression under depression and provided a novel strategy for GC-mediated antidepressant therapy in patients with liver cancer.
Collapse
Affiliation(s)
- Yawei Zhao
- Department of Pharmacology, College of Basic Medical Sciences
| | - Yong Jia
- School of Nursing, Jilin University, Changchun, China
| | - Tongfei Shi
- Department of Pharmacology, College of Basic Medical Sciences
| | - Wencong Wang
- Department of Pharmacology, College of Basic Medical Sciences
| | - Dan Shao
- Department of Pharmacology, College of Basic Medical Sciences
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Xiao Zheng
- Department of Pharmacology, College of Basic Medical Sciences
| | - Madi Sun
- Department of Pharmacology, College of Basic Medical Sciences
| | - Kan He
- Department of Pharmacology, College of Basic Medical Sciences
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences
- School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
63
|
Pan EY, Merl MY, Lin K. The impact of corticosteroid use during anti-PD1 treatment. J Oncol Pharm Pract 2019; 26:814-822. [DOI: 10.1177/1078155219872786] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background The advent of anti-PD1 therapy for cancer treatment has led to improvements in response rates and overall survival. However, anti-PD1 therapy has the potential to cause immune-related adverse events (irAEs), which can be treated with corticosteroids if severe. The clinical implications of concomitant immunotherapy and systemic steroids remain unclear, as short courses of steroids do not significantly suppress T-cell function. The primary objective of this study is to determine if the use of concomitant steroids impacts the efficacy of anti-PD1 therapy. Methods This retrospective, single-center study reviewed adult patients who received at least four cycles of nivolumab or pembrolizumab for the treatment of melanoma, non-small cell lung cancer (NSCLC), or renal cell carcinoma from November 2014 to February 2016. Patients who received steroids (prednisone equivalent >10 mg) during anti-PD1 therapy were divided into two main cohorts based on the duration of steroid administration of ≤2 weeks or >2 weeks. Time to disease progression, overall response, and overall survival were assessed. Results Twenty-seven of 55 patients (13 melanoma, 11 NSCLC, 3 renal cell carcinoma) required steroids during anti-PD1 therapy. In patients who received steroids, median time to disease progression was 5.6 months for melanoma, 5.8 for NSCLC, and 2.0 for renal cell carcinoma. The overall response rate (ORR) was 3/13 (23%) for melanoma, 6/11 (54%) for NSCLC, and 1/3 (33%) for renal cell carcinoma. Median overall survival was 11.9 months for melanoma, 9.9 for NSCLC, and not reached for renal cell carcinoma. Thirteen patients who had received steroids expired; 11 of these patients had received prednisone >10 mg/day for >2 weeks. Conclusion High-dose steroids for long durations during anti-PD1 therapy may be associated with poorer survival outcomes.
Collapse
Affiliation(s)
- Eva Y Pan
- Smilow Cancer Hospital of Yale-New Haven Health, New Haven, CT, USA
| | - Man Yee Merl
- Smilow Cancer Hospital of Yale-New Haven Health, New Haven, CT, USA
| | - Katherine Lin
- Smilow Cancer Hospital of Yale-New Haven Health, New Haven, CT, USA
| |
Collapse
|
64
|
Agina HA, Ehsan NA, Abd-Elaziz TA, Abd-Elfatah GA, Said EM, Sira MM. Hepatic expression of programmed death-1 (PD-1) and its ligand, PD-L1, in children with autoimmune hepatitis: relation to treatment response. Clin Exp Hepatol 2019; 5:256-264. [PMID: 31598564 PMCID: PMC6781821 DOI: 10.5114/ceh.2019.87642] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 06/01/2019] [Indexed: 02/07/2023] Open
Abstract
AIM OF THE STUDY Autoimmune hepatitis (AIH) is characterized histologically by aggressive inflammation with interface hepatitis and prominent lymphoplasmacytic infiltration. Programmed death-1 (PD-1) is expressed on activated lymphocytes. Engagement of PD-1 by its ligand PD-L1 leads to cell apoptosis and death. We aimed to evaluate the immunohistochemical expression of PD-1 and PD-L1 in children with AIH, and its relation to treatment outcome. MATERIAL AND METHODS Pre-treatment liver biopsies of 31 children with AIH were compared to 30 children with chronic hepatitis C virus (HCV) infection as a control group. PD-1 was evaluated in lymphocytes, while PD-L1 was evaluated in lymphocytes, hepatocytes, biliary epithelial cells, sinusoidal endothelial cells and Kupffer cells. All AIH patients received the standard treatment. RESULTS The mean PD-1 was significantly higher in AIH than HCV patients (29.19 ±18.5% vs. 15.2 ±10.1%; p = 0.002) while there was no statistically significant difference as regards PD-L1 on lymphocytes (p = 0.853). Neither PD-1 nor PD-L1 correlated with either liver fibrosis or the inflammatory activity (p > 0.05 for all). PD-1/PD-L1 ratio was significantly higher in AIH compared to HCV patients and in non-responder AIH patients compared to responders (46.9 vs. 6.58). PD-1 expression was comparable in both responders and non-responders (p = 0.813), while PD-L1 was significantly upregulated in responders (4.17 ±3.15% vs. 0.63 ±1.3%; p = 0.046). PD-L1 expression on hepatocytes, biliary epithelial cells, sinusoidal endothelial cells and Kupffer cells was comparable in AIH and HCV groups. CONCLUSIONS PD-1/PD-L1 ratio, which reflects immune aggression, was significantly higher in AIH compared to HCV patients and in non-responder AIH patients compared to responders.
Collapse
Affiliation(s)
- Hala A Agina
- Pathology Department, Faculty of Medicine, Benha University, Egypt
| | - Nermine A Ehsan
- Pathology Department, National Liver Institute, Menoufia University, Egypt
| | | | | | - Eman M Said
- Pathology Department, Faculty of Medicine, Benha University, Egypt
| | - Mostafa M Sira
- Pediatric Hepatology, Gastroenterology and Nutrition Department, National Liver Institute, Menoufia University, Egypt
| |
Collapse
|
65
|
Rudak PT, Gangireddy R, Choi J, Burhan AM, Summers KL, Jackson DN, Inoue W, Haeryfar SMM. Stress-elicited glucocorticoid receptor signaling upregulates TIGIT in innate-like invariant T lymphocytes. Brain Behav Immun 2019; 80:793-804. [PMID: 31108170 DOI: 10.1016/j.bbi.2019.05.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/22/2019] [Accepted: 05/16/2019] [Indexed: 12/17/2022] Open
Abstract
Stress is known to impede certain host defense mechanisms, including those governed by conventional T lymphocytes. However, whether innate-like T lymphocytes, such as invariant natural killer T (iNKT) and mucosa-associated invariant T (MAIT) cells, are impacted by stress is unclear. Herein, we report that prolonged psychological stress caused by physical confinement results in robust upregulation of T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), an immune checkpoint receptor that controls antitumor and antiviral immune responses. Elevated TIGIT expression was found not only on NK and conventional T cells, but also on iNKT and MAIT cells. Stress-provoked TIGIT upregulation was reversed through treatment with the glucocorticoid receptor (GR) antagonist RU486, but not with 6-hydroxydopamine that induces chemical sympathectomy. A Cre/Lox gene targeting model in which GR was ablated in cells expressing Lck under its proximal promoter revealed that TIGIT upregulation in stressed animals stems from direct GR signaling in T and iNKT cells. In fact, long-term oral administration of exogenous corticosterone (CS) to wild-type C57BL/6 (B6) mice was sufficient to increase TIGIT expression levels on T and iNKT cells. In vitro treatment with CS also potently and selectively upregulated TIGIT, but not CTLA-4 or LAG-3, on mouse iNKT and MAIT hybridomas. These results were recapitulated using primary hepatic iNKT and MAIT cells from wild-type B6 and B6.MAITCAST mice, respectively. Subjecting B6.MAITCAST mice to physical restraint also raised the frequency of TIGIT+ cells among hepatic MAIT cells in a GR-dependent manner. Finally, we found that TIGIT is similarly upregulated in a chronic variable stress model in which animals are exposed to unpredictable heterotypic stressors without developing habituation. Taken together, our findings link, for the first time to our knowledge, GR signaling to TIGIT expression. We propose that glucocorticoid hormones dampen immune responses, in part, by enhancing TIGIT expression across multiple critical subsets of effector lymphocytes, including innate-like T cells. Therefore, TIGIT may constitute an attractive target in immune-enhancing interventions for sustained physiological stress.
Collapse
Affiliation(s)
- Patrick T Rudak
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Rakshith Gangireddy
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Joshua Choi
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Amer M Burhan
- Department of Psychiatry, Western University, London, Ontario, Canada
| | - Kelly L Summers
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Dwayne N Jackson
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Wataru Inoue
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Department of Medicine, Division of Clinical Immunology and Allergy, Western University, London, Ontario, Canada; Department of Surgery, Division of General Surgery, Western University, London, Ontario, Canada.
| |
Collapse
|
66
|
Aston WJ, Hope DE, Cook AM, Boon L, Dick I, Nowak AK, Lake RA, Lesterhuis WJ. Dexamethasone differentially depletes tumour and peripheral blood lymphocytes and can impact the efficacy of chemotherapy/checkpoint blockade combination treatment. Oncoimmunology 2019; 8:e1641390. [PMID: 31646089 PMCID: PMC6791454 DOI: 10.1080/2162402x.2019.1641390] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 10/26/2022] Open
Abstract
Dexamethasone is a synthetic glucocorticoid commonly used for the prevention and management of side effects in cancer patients undergoing chemotherapy. While it is effective as an anti-emetic and in preventing hypersensitivity reactions, dexamethasone depletes peripheral blood lymphocytes and impacts immune responses. The effect of dexamethasone on the number and quality of tumour-infiltrating leukocytes has not been reported. To address this, we calibrated the dose in two different strains of mice to achieve the same extent of peripheral blood lymphocyte depletion observed in patients with cancer. Doses that caused analogous depletion of T and B lymphocytes and NK cells from the peripheral blood, elicited no change in these populations within the tumour. The expression of immune checkpoint molecules PD-1, OX40, GITR and TIM3 on tumour-infiltrating lymphocytes was not altered. We found that dexamethasone had a small but significant deleterious impact on weakly efficacious chemoimmunotherapy but had no effect when the protocol was highly efficacious. Based on these results, we predict that dexamethasone will have a modest negative influence on the overall effectiveness of chemoimmunotherapy treatment.
Collapse
Affiliation(s)
- Wayne J. Aston
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- Medical School, The University of Western Australia, Nedlands, WA, Australia
| | - Danika E. Hope
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Alistair M. Cook
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- Medical School, The University of Western Australia, Nedlands, WA, Australia
| | | | - Ian Dick
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Anna K. Nowak
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- Medical School, The University of Western Australia, Nedlands, WA, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Richard A. Lake
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - W. Joost Lesterhuis
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
67
|
Wofford KL, Cullen DK, Spiller KL. Modulation of macrophage phenotype via phagocytosis of drug-loaded microparticles. J Biomed Mater Res A 2019; 107:1213-1224. [PMID: 30672109 PMCID: PMC6499658 DOI: 10.1002/jbm.a.36617] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 01/09/2019] [Indexed: 12/17/2022]
Abstract
Monocyte-derived macrophages play a critical role in directing wound pathology following injury. Depending on their phenotype, macrophages also promote tissue regeneration. However, the therapeutic administration of macrophages with a controlled phenotype is challenging because macrophages are highly plastic and quickly revert to a detrimental, inflammatory phenotype in response to the environment of a damaged tissue. To address this issue, we developed a novel strategy to modulate macrophage phenotype intracellularly through phagocytosis of drug-loaded microparticles. Poly(lactic-co-glycolic acid) microparticles loaded with the anti-inflammatory drug dexamethasone (Dex) were phagocytosed by monocytes and stored intracellularly for at least 5 days. After differentiation into macrophages, cell phenotype was characterized over time with high-throughput gene expression analysis via NanoString. We found that the microparticles modulated macrophage phenotype for up to 7 days after microparticle uptake, with decreases in inflammation-related genes at early timepoints and upregulation of homing- and phagocytosis-related genes at multiple timepoints in a manner similar to cells treated with continuous free Dex. These data suggest that intracellularly loading macrophages with Dex microparticles via phagocytosis could be a unique methodology to selectively modulate macrophage phenotype over time. This strategy would allow therapeutic administration of macrophages for the treatment of a number of inflammatory disease and disorders. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1213-1224, 2019.
Collapse
Affiliation(s)
- Kathryn L Wofford
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania
- Center for Neurotrauma, Neurodegeneration and Restoration, CMC VA Medical Center, Philadelphia, Pennsylvania
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - D Kacy Cullen
- Center for Neurotrauma, Neurodegeneration and Restoration, CMC VA Medical Center, Philadelphia, Pennsylvania
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kara L Spiller
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania
| |
Collapse
|
68
|
Jain S, Dash P, Minz AP, Satpathi S, Samal AG, Behera PK, Satpathi PS, Senapati S. Lipopolysaccharide (LPS) enhances prostate cancer metastasis potentially through NF-κB activation and recurrent dexamethasone administration fails to suppress it in vivo. Prostate 2019; 79:168-182. [PMID: 30264470 DOI: 10.1002/pros.23722] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 09/13/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Previous studies have shown the effect of bacterial lipopolysaccharide (LPS) on enhanced cancer cells' growth and metastasis. However, the effect of LPS on prostate cancer (PCa) cells metastasis has not been investigated in details. This study aimed to investigate the functional role of LPS on PCa cells metastasis and determine the effect of dexamethasone (DEX) on this event. METHODS Two different PCa reporter cells lines (DU145-NF-κB-Luc and MAT-LyLu- NF-κB-Luc) were used to assess the direct effect of LPS on NF-κB activation in PCa cells. Plasma collected from LPS-stimulated human and rodent blood were used to check the indirect effect of LPS on NF-κB activation in PCa cells. Trans-well migration assay and two different orthotopic PCa animal models were used to investigate the effect of LPS on DU145 and MAT-LyLu cells migration or metastasis in vitro and in vivo, respectively. In all the studies DEX was used with or without LPS stimulation. RESULTS LPS and secretory factors present in plasma collected from LPS-stimulated blood, significantly activated NF-κB in DU145, and MAT-LyLu cells and enhanced their migration in vitro. DEX significantly suppressed LPS-mediated activation of cancer and blood cells and abrogated the direct and indirect pro-migratory effect of LPS on PCa cells. Systemic administration of LPS activated NF-κB in DU145 cells in vivo; however, failed to alter the metastatic properties of these cells. On the other hand, systemic administration of LPS to MAT-LyLu tumor bearing animals significantly enhanced the incidence of metastasis without altering the overall growth of primary tumors. Unexpectedly, though DEX significantly suppressed MAT-LyLu primary tumor weights, it aggravated metastasis of cancer cells in presence and absence of LPS. Moreover, consecutive DEX pre-treatment enhanced experimental peritoneal metastasis of MAT-LyLu cells. At the molecular level, LPS, and/or DEX induced overexpression of immunosuppressive molecules in MAT-LyLu tumors. CONCLUSIONS Overall, our study has shown that LPS and/or LPS induced inflammation can increase PCa metastasis and immunosuppressive dose of DEX might further enhance cancer metastasis.
Collapse
Affiliation(s)
- Sumeet Jain
- Tumor Microenvironment and Animal Models Lab, Institute of Life Sciences, Bhubaneswar, Odisha, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Pujarini Dash
- Tumor Microenvironment and Animal Models Lab, Institute of Life Sciences, Bhubaneswar, Odisha, India
| | - Aliva P Minz
- Tumor Microenvironment and Animal Models Lab, Institute of Life Sciences, Bhubaneswar, Odisha, India
| | | | - Ajit G Samal
- Department of Surgery, Hitech Medical College, Rourkela, Odisha, India
| | - Prativa K Behera
- Department of Pathology, Ispat General Hospital, Rourkela, Odisha, India
| | - Partha S Satpathi
- Department of Microbiology, Midnapore Medical College, Midnapore, West Bengal, India
| | - Shantibhusan Senapati
- Tumor Microenvironment and Animal Models Lab, Institute of Life Sciences, Bhubaneswar, Odisha, India
| |
Collapse
|
69
|
Taha T, Meiri D, Talhamy S, Wollner M, Peer A, Bar-Sela G. Cannabis Impacts Tumor Response Rate to Nivolumab in Patients with Advanced Malignancies. Oncologist 2019; 24:549-554. [PMID: 30670598 DOI: 10.1634/theoncologist.2018-0383] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 12/11/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND There has been a significant increase in the use of immunotherapy and cannabis recently, two modalities that have immunomodulatory effects and may have possible interaction. We evaluated the influence of cannabis use during immunotherapy treatment on response rate (RR), progression-free survival (PFS), and overall survival (OS). SUBJECTS, MATERIALS, AND METHODS In this retrospective, observational study, data were collected from the files of patients treated with nivolumab in the years 2015-2016 at our hospital, and cannabis from six cannabis-supplying companies. Included were 140 patients (89 nivolumab alone, 51 nivolumab plus cannabis) with advanced melanoma, non-small cell lung cancer, and renal clear cell carcinoma. The groups were homogenous regarding demographic and disease characteristics. A comparison between the two arms was made. RESULTS In a multivariate model, cannabis was the only significant factor that reduced RR to immunotherapy (37.5% RR in nivolumab alone compared with 15.9% in the nivolumab-cannabis group (p = .016, odds ratio = 3.13, 95% confidence interval 1.24-8.1). Cannabis use was not a significant factor for PFS or OS. Factors affecting PFS and OS were smoking (adjusted hazard ratio [HR] = 2.41 and 2.41, respectively (and brain metastases (adjusted HR = 2.04 and 2.83, respectively). Low performance status (adjusted HR = 2.83) affected OS alone. Tetrahydrocannabinol and cannabidiol percentages did not affect RR in any group (p = .393 and .116, respectively). CONCLUSION In this retrospective analysis, the use of cannabis during immunotherapy treatment decreased RR, without affecting PFS or OS and without relation to cannabis composition. Considering the limitations of the study, further prospective clinical study is needed to investigate possible interaction. IMPLICATIONS FOR PRACTICE Although the data are retrospective and a relation to cannabis composition was not detected, this information can be critical for cannabis users and indicates that caution is required when starting immunotherapy.
Collapse
Affiliation(s)
- Tarek Taha
- Division of Oncology, Rambam Health Care Campus, Haifa, Israel
| | - David Meiri
- Laboratory of Cancer Biology and Cannabinoids Research, Department of Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Samira Talhamy
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Mira Wollner
- Division of Oncology, Rambam Health Care Campus, Haifa, Israel
| | - Avivit Peer
- Division of Oncology, Rambam Health Care Campus, Haifa, Israel
| | - Gil Bar-Sela
- Division of Oncology, Rambam Health Care Campus, Haifa, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
70
|
Autoimmune disease-associated non-Hodgkin’s lymphoma—a large retrospective study from China. Ann Hematol 2018; 98:445-455. [DOI: 10.1007/s00277-018-3515-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 10/04/2018] [Indexed: 12/19/2022]
|
71
|
Altwerger G, Florsheim EB, Menderes G, Black J, Schwab C, Gressel GM, Nelson WK, Carusillo N, Passante T, Huang G, Litkouhi B, Azodi M, Silasi DA, Santin A, Schwartz PE, Ratner ES. Impact of carboplatin hypersensitivity and desensitization on patients with recurrent ovarian cancer. J Cancer Res Clin Oncol 2018; 144:2449-2456. [DOI: 10.1007/s00432-018-2753-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 09/11/2018] [Indexed: 01/31/2023]
|
72
|
de Goeje PL, Klaver Y, Kaijen-Lambers MEH, Langerak AW, Vroman H, Kunert A, Lamers CHJ, Aerts JGJV, Debets R, Hendriks RW. Autologous Dendritic Cell Therapy in Mesothelioma Patients Enhances Frequencies of Peripheral CD4 T Cells Expressing HLA-DR, PD-1, or ICOS. Front Immunol 2018; 9:2034. [PMID: 30245692 PMCID: PMC6137618 DOI: 10.3389/fimmu.2018.02034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/17/2018] [Indexed: 01/05/2023] Open
Abstract
Introduction: Malignant pleural mesothelioma (MPM) is a malignancy with a very poor prognosis for which new treatment options are urgently needed. We have previously shown that dendritic cell (DC) immunotherapy provides a clinically feasible treatment option. In the current study, we set out to assess the immunological changes induced by DC immunotherapy in peripheral blood of MPM patients. Methods: Peripheral blood was collected from nine patients enrolled in a phase I dose escalation study, before and after treatment with DCs that were pulsed with an allogeneic tumor lysate preparation consisting of a mixture of five cultured mesothelioma cell lines. We used immune profiling by multiplex flow cytometry to characterize different populations of immune cells. In particular, we determined frequencies of T cell subsets that showed single and combinatorial expression of multiple markers that signify T cell activation, maturation and inhibition. Therapy-induced T cell reactivity was assessed in peptide/MHC multimer stainings using mesothelin as a prototypic target antigen with confirmed expression in the clinical tumor lysate preparation. T cell receptor (TCR) diversity was evaluated by TCRB gene PCR assays. Results: We observed an increase in the numbers of B cells, CD4 and CD8 T cells, but not NK cells at 6 weeks post-treatment. The increases in B and T lymphocytes were not accompanied by major changes in T cell reactivity toward mesothelin nor in TCRB diversity. Notably, we did observe enhanced proportions of CD4 T cells expressing HLA-DR, PD-1 (at 2 weeks after onset of treatment) and ICOS (6 weeks) and a CD8 T cell population expressing LAG3 (2 weeks). Discussion: DC immunotherapy using allogeneic tumor lysate resulted in enhanced frequencies of B cells and T cells in blood. We did not detect a skewed antigen-reactivity of peripheral CD8 T cells. Interestingly, frequencies of CD4 T cells expressing activation markers and PD-1 were increased. These findings indicate a systemic activation of the adaptive immune response and may guide future immune monitoring studies of DC therapies.
Collapse
Affiliation(s)
- Pauline L de Goeje
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Yarne Klaver
- Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, Netherlands.,Department of Medical Oncology, Laboratory of Tumor Immunology, Erasmus MC, Rotterdam, Netherlands
| | - Margaretha E H Kaijen-Lambers
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Anton W Langerak
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, Rotterdam, Netherlands
| | - Heleen Vroman
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - André Kunert
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, Netherlands.,Department of Medical Oncology, Laboratory of Tumor Immunology, Erasmus MC, Rotterdam, Netherlands
| | - Cor H J Lamers
- Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, Netherlands.,Department of Medical Oncology, Laboratory of Tumor Immunology, Erasmus MC, Rotterdam, Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Reno Debets
- Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, Netherlands.,Department of Medical Oncology, Laboratory of Tumor Immunology, Erasmus MC, Rotterdam, Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
73
|
Rew Y, Du X, Eksterowicz J, Zhou H, Jahchan N, Zhu L, Yan X, Kawai H, McGee LR, Medina JC, Huang T, Chen C, Zavorotinskaya T, Sutimantanapi D, Waszczuk J, Jackson E, Huang E, Ye Q, Fantin VR, Sun D. Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101). J Med Chem 2018; 61:7767-7784. [PMID: 30091920 DOI: 10.1021/acs.jmedchem.8b00743] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The glucocorticoid receptor (GR) has been linked to therapy resistance across a wide range of cancer types. Preclinical data suggest that antagonists of this nuclear receptor may enhance the activity of anticancer therapy. The first-generation GR antagonist mifepristone is currently undergoing clinical evaluation in various oncology settings. Structure-based modification of mifepristone led to the discovery of ORIC-101 (28), a highly potent steroidal GR antagonist with reduced androgen receptor (AR) agonistic activity amenable for dosing in androgen receptor positive tumors and with improved CYP2C8 and CYP2C9 inhibition profile to minimize drug-drug interaction potential. Unlike mifepristone, 28 could be codosed with chemotherapeutic agents readily metabolized by CYP2C8 such as paclitaxel. Furthermore, 28 demonstrated in vivo antitumor activity by enhancing response to chemotherapy in the GR+ OVCAR5 ovarian cancer xenograft model. Clinical evaluation of safety and therapeutic potential of 28 is underway.
Collapse
Affiliation(s)
- Yosup Rew
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Xiaohui Du
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - John Eksterowicz
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Haiying Zhou
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Nadine Jahchan
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Liusheng Zhu
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Xuelei Yan
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Hiroyuki Kawai
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Lawrence R McGee
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Julio C Medina
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Tom Huang
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Chelsea Chen
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Tatiana Zavorotinskaya
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Dena Sutimantanapi
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Joanna Waszczuk
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Erica Jackson
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Elizabeth Huang
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Qiuping Ye
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Valeria R Fantin
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| | - Daqing Sun
- ORIC Pharmaceuticals , 240 East Grand Avenue, Fl2 , South San Francisco , California 94080 , United States
| |
Collapse
|
74
|
Murine Models of Steroid Refractory Graft-versus-Host Disease. Sci Rep 2018; 8:12475. [PMID: 30127532 PMCID: PMC6102256 DOI: 10.1038/s41598-018-30814-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/06/2018] [Indexed: 12/20/2022] Open
Abstract
Corticosteroids are the first line therapy for acute graft-versus-host disease (GVHD). However, the outcome of steroid refractory GVHD (SR-GVHD) is poor due to a lack of effective treatments. The development of therapies for SR-GVHD is limited by an incomplete understanding of its pathophysiology partly because of the absence of clinically relevant animal models of SR-GVHD. Here we addressed the need for a SR-GVHD animal model by developing both MHC matched multiple minor histocompatibility antigens (miHAs) mismatched and MHC mismatched haploidentical murine models of SR-GVHD. We demonstrate that animals can develop SR-GVHD regardless of whether steroids are initiated early or late post allogeneic bone marrow transplantation (allo-BMT). In general, we observed increased GVHD specific histopathological damage of target organs in SR-GVHD animals relative to steroid responsive animals. Interestingly, we found no significant differences in donor T cell characteristics between steroid refractory and responsive animals suggesting that donor T cell independent mechanisms may play more prominent roles in the pathogenesis of SR-GVHD than was considered previously.
Collapse
|
75
|
Banna GL, Passiglia F, Colonese F, Canova S, Menis J, Addeo A, Russo A, Cortinovis DL. Immune-checkpoint inhibitors in non-small cell lung cancer: A tool to improve patients' selection. Crit Rev Oncol Hematol 2018; 129:27-39. [PMID: 30097235 DOI: 10.1016/j.critrevonc.2018.06.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 06/16/2018] [Accepted: 06/18/2018] [Indexed: 12/15/2022] Open
Abstract
The identification of reliable predictive biomarkers of efficacy or resistance to immune-oncology (I-O) agents is a major issue for translational research and clinical practice. However, along with PDL1 and molecular features other clinical, radiological and laboratory factors can be considered for the selection of those patients who would not be the best candidate for immune-checkpoint inhibitors (ICPIs). We examined these factors, emerging from the results of currently available studies in non-small cell lung cancer (NSCLC), aiming to provide a useful and manageable tool which can help Oncologists in their everyday clinical practice. A thorough patient evaluation and close clinical monitoring, due to limited, early or inconclusive currently available data, should be deserved for patients with a pre-existing symptomatic chronic obstructive pulmonary disease, age >75 years, Eastern Cooperative Oncology Group (ECOG) performance status (PS) ≥ 1, a time to progression (TTP) < three months and progressive disease (PD) as the best response to the previous treatment, hepatitis or HIV-infections, high neutrophil to lymphocyte ratio (NLR), or on treatment with high-dose steroids, when the use of ICPIs is considered. Limited data are available to consider that ICPIs are safe in patients with interstitial lung disease, bronchiolitis obliterans organizing pneumonia and autommune diseases. Early evidence on steroids, vaccinations and antibiotics suggest their possible interaction with ICPIs and need to be more investigated in clinical trials. Oncogene-addicted NSCLC harboring EGFR-mutations and low tumor-infiltrating T-lymphocytes (TILs) seems not to gain benefit from I-O.
Collapse
Affiliation(s)
- Giuseppe Luigi Banna
- Division of Medical Oncology, Cannizzaro Hospital, Via Messina 829, 95126, Catania, Italy.
| | - Francesco Passiglia
- Department of Surgical, Oncological and Stomatological Disciplines, University of Palermo, Italy
| | | | | | - Jessica Menis
- Department of Oncology Medicine, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Alfredo Addeo
- Oncology Department, University Hospital Geneva, 1205 Geneva, Switzerland
| | - Antonio Russo
- Department of Surgical, Oncological and Stomatological Disciplines, University of Palermo, Italy
| | | |
Collapse
|
76
|
Giles AJ, Hutchinson MKND, Sonnemann HM, Jung J, Fecci PE, Ratnam NM, Zhang W, Song H, Bailey R, Davis D, Reid CM, Park DM, Gilbert MR. Dexamethasone-induced immunosuppression: mechanisms and implications for immunotherapy. J Immunother Cancer 2018; 6:51. [PMID: 29891009 PMCID: PMC5996496 DOI: 10.1186/s40425-018-0371-5] [Citation(s) in RCA: 305] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/30/2018] [Indexed: 12/18/2022] Open
Abstract
Background Corticosteroids are routinely utilized to alleviate edema in patients with intracranial lesions and are first-line agents to combat immune-related adverse events (irAEs) that arise with immune checkpoint blockade treatment. However, it is not known if or when corticosteroids can be administered without abrogating the efforts of immunotherapy. The purpose of this study was to evaluate the impact of dexamethasone on lymphocyte activation and proliferation during checkpoint blockade to provide guidance for corticosteroid use while immunotherapy is being implemented as a cancer treatment. Methods Lymphocyte proliferation, differentiation, and cytokine production were evaluated during dexamethasone exposure. Human T cells were stimulated through CD3 ligation and co-stimulated either directly by CD28 ligation or by providing CD80, a shared ligand for CD28 and CTLA-4. CTLA-4 signaling was inhibited by antibody blockade using ipilimumab which has been approved for the treatment of several solid tumors. The in vivo effects of dexamethasone during checkpoint blockade were evaluated using the GL261 syngeneic mouse intracranial model, and immune populations were profiled by flow cytometry. Results Dexamethasone upregulated CTLA-4 mRNA and protein in CD4 and CD8 T cells and blocked CD28-mediated cell cycle entry and differentiation. Naïve T cells were most sensitive, leading to a decrease of the development of more differentiated subsets. Resistance to dexamethasone was conferred by blocking CTLA-4 or providing strong CD28 co-stimulation prior to dexamethasone exposure. CTLA-4 blockade increased IFNγ expression, but not IL-2, in stimulated human peripheral blood T cells exposed to dexamethasone. Finally, we found that CTLA-4 blockade partially rescued T cell numbers in mice bearing intracranial gliomas. CTLA-4 blockade was associated with increased IFNγ-producing tumor-infiltrating T cells and extended survival of dexamethasone-treated mice. Conclusions Dexamethasone-mediated T cell suppression diminishes naïve T cell proliferation and differentiation by attenuating the CD28 co-stimulatory pathway. However, CTLA-4, but not PD-1 blockade can partially prevent some of the inhibitory effects of dexamethasone on the immune response. Electronic supplementary material The online version of this article (10.1186/s40425-018-0371-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Amber J Giles
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA.
| | - Marsha-Kay N D Hutchinson
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Heather M Sonnemann
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Jinkyu Jung
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Peter E Fecci
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Nivedita M Ratnam
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Wei Zhang
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Hua Song
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Rolanda Bailey
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Dionne Davis
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Caitlin M Reid
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Deric M Park
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| |
Collapse
|
77
|
Ayroldi E, Cannarile L, Delfino DV, Riccardi C. A dual role for glucocorticoid-induced leucine zipper in glucocorticoid function: tumor growth promotion or suppression? Cell Death Dis 2018; 9:463. [PMID: 29695779 PMCID: PMC5916931 DOI: 10.1038/s41419-018-0558-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/27/2018] [Accepted: 03/30/2018] [Indexed: 02/06/2023]
Abstract
Glucocorticoids (GCs), important therapeutic tools to treat inflammatory and immunosuppressive diseases, can also be used as part of cancer therapy. In oncology, GCs are used as anticancer drugs for lymphohematopoietic malignancies, while in solid neoplasms primarily to control the side effects of chemo/radiotherapy treatments. The molecular mechanisms underlying the effects of GCs are numerous and often overlapping, but not all have been elucidated. In normal, cancerous, and inflammatory tissues, the response to GCs differs based on the tissue type. The effects of GCs are dependent on several factors: the tumor type, the GC therapy being used, the expression level of the glucocorticoid receptor (GR), and the presence of any other stimuli such as signals from immune cells and the tumor microenvironment. Therefore, GCs may either promote or suppress tumor growth via different molecular mechanisms. Stress exposure results in dysregulation of the hypothalamic-pituitary-adrenal axis with increased levels of endogenous GCs that promote tumorigenesis, confirming the importance of GCs in tumor growth. Most of the effects of GCs are genomic and mediated by the modulation of GR gene transcription. Moreover, among the GR-induced genes, glucocorticoid-induced leucine zipper (GILZ), which was cloned and characterized primarily in our laboratory, mediates many GC anti-inflammatory effects. In this review, we analyzed the possible role for GILZ in the effects GCs have on tumors cells. We also suggest that GILZ, by affecting the immune system, tumor microenvironment, and directly cancer cell biology, has a tumor-promoting function. However, it may also induce apoptosis or decrease the proliferation of cancer cells, thus inhibiting tumor growth. The potential therapeutic implications of GILZ activity on tumor cells are discussed here.
Collapse
Affiliation(s)
- Emira Ayroldi
- Department of Medicine, Section of Pharmacology, Medical School, University of Perugia, Perugia, Italy.
| | - Lorenza Cannarile
- Department of Medicine, Section of Pharmacology, Medical School, University of Perugia, Perugia, Italy
| | - Domenico V Delfino
- Department of Medicine, Section of Pharmacology, Medical School, University of Perugia, Perugia, Italy
| | - Carlo Riccardi
- Department of Medicine, Section of Pharmacology, Medical School, University of Perugia, Perugia, Italy
| |
Collapse
|
78
|
Boussiotis VA, Charest A. Immunotherapies for malignant glioma. Oncogene 2018; 37:1121-1141. [PMID: 29242608 PMCID: PMC5828703 DOI: 10.1038/s41388-017-0024-z] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 12/31/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant primary brain cancer with a dreadful overall survival and for which treatment options are limited. Recent breakthroughs in novel immune-related treatment strategies for cancer have spurred interests in usurping the power of the patient's immune system to recognize and eliminate GBM. Here, we discuss the unique properties of GBM's tumor microenvironment, the effects of GBM standard on care therapy on tumor-associated immune cells, and review several approaches aimed at therapeutically targeting the immune system for GBM treatment. We believe that a comprehensive understanding of the intricate micro-environmental landscape of GBM will abound into the development of novel immunotherapy strategies for GBM patients.
Collapse
Affiliation(s)
- Vassiliki A Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Alain Charest
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA, USA.
- Division of Genetics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
79
|
Takahashi T, Hsiao HM, Tanaka S, Li W, Higashikubo R, Scozzi D, Bharat A, Ritter JH, Krupnick AS, Gelman AE, Kreisel D. PD-1 expression on CD8 + T cells regulates their differentiation within lung allografts and is critical for tolerance induction. Am J Transplant 2018; 18:216-225. [PMID: 28730633 PMCID: PMC5739961 DOI: 10.1111/ajt.14437] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/18/2017] [Accepted: 07/11/2017] [Indexed: 01/25/2023]
Abstract
Immunological requirements for rejection and tolerance induction differ between various organs. While memory CD8+ T cells are considered a barrier to immunosuppression-mediated acceptance of most tissues and organs, tolerance induction after lung transplantation is critically dependent on central memory CD8+ T lymphocytes. Here we demonstrate that costimulation blockade-mediated tolerance after lung transplantation is dependent on programmed cell death 1 (PD-1) expression on CD8+ T cells. In the absence of PD-1 expression, CD8+ T cells form prolonged interactions with graft-infiltrating CD11c+ cells; their differentiation is skewed towards an effector memory phenotype and grafts are rejected acutely. These findings extend the notion that requirements for tolerance induction after lung transplantation differ from other organs. Thus, immunosuppressive strategies for lung transplant recipients need to be tailored based on the unique immunological properties of this organ.
Collapse
Affiliation(s)
- T Takahashi
- Department of Surgery, Washington University in St. Louis
| | - HM Hsiao
- Department of Surgery, Washington University in St. Louis
| | - S Tanaka
- Department of Surgery, Washington University in St. Louis
| | - W Li
- Department of Surgery, Washington University in St. Louis
| | - R Higashikubo
- Department of Surgery, Washington University in St. Louis
| | - D Scozzi
- Department of Surgery, Washington University in St. Louis
| | - A Bharat
- Department of Surgery, Northwestern University
| | - JH Ritter
- Department of Pathology & Immunology, Washington University in St. Louis
| | - AS Krupnick
- Department of Surgery, University of Virginia
| | - AE Gelman
- Department of Surgery, Washington University in St. Louis,Department of Pathology & Immunology, Washington University in St. Louis
| | - D Kreisel
- Department of Surgery, Washington University in St. Louis,Department of Pathology & Immunology, Washington University in St. Louis,Corresponding author: Daniel Kreisel, M.D., Ph.D., Professor of Surgery, Pathology & Immunology, Campus Box 8234, 660 South Euclid Avenue, Washington University School of Medicine, St. Louis, MO 63110, Tel: (314) 362-6021, Fax: (314) 367-8459,
| |
Collapse
|
80
|
Garant A, Guilbault C, Ekmekjian T, Greenwald Z, Murgoi P, Vuong T. Concomitant use of corticosteroids and immune checkpoint inhibitors in patients with hematologic or solid neoplasms: A systematic review. Crit Rev Oncol Hematol 2017; 120:86-92. [DOI: 10.1016/j.critrevonc.2017.10.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/22/2017] [Indexed: 12/17/2022] Open
|
81
|
Neutrophil to Lymphocyte Ratio in Castration-Resistant Prostate Cancer Patients Treated With Daily Oral Corticosteroids. Clin Genitourin Cancer 2017; 15:678-684.e1. [PMID: 28606735 DOI: 10.1016/j.clgc.2017.05.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/28/2017] [Accepted: 05/03/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND The neutrophil to lymphocyte ratio (NLR) has been shown to be highly prognostic across many tumor types, and predictive of treatment outcome in advanced prostate cancer, and has been postulated to be an indirect measure of tumor inflammation. We evaluated the effect of low-dose steroids on NLR in men suffering from castration-resistant prostate cancer (CRPC). PATIENTS AND METHODS The NLR was evaluated in a prospective randomized phase II trial that compared prednisolone 5 mg twice daily and dexamethasone 0.5 mg daily administered to 75 chemotherapy and abiraterone/enzalutamide-naive CRPC patients. NLR was examined at baseline (BL), after 6 and 12 weeks of corticosteroid treatment; associations with >50% prostate-specific antigen (PSA) response, duration of response (PSA progression-free interval), and overall survival (OS) were tested using logistic regression and Cox regression analysis. RESULTS The median NLR for all evaluable patients was 2.6 at BL; 2.9 at 6 weeks; and 4.0 at 12 weeks. After low-dose corticosteroid initiation, 46 patients had a decline in PSA with 24 confirmed responders. BL NLR (log10) associated with a PSA response (odds ratio, .029, 95% confidence interval [CI], .002-.493; P = .014), and with the extent of the PSA decline (P = .009). A favorable BL NLR (less than median) associated with a 5.5-fold higher odds of a PSA >50% response (95% CI, 1.3-23.9; P = .02). Higher BL NLR (log10) associated with a shorter time to PSA progression (hazard ratio [HR], 9.5; 95% CI, 2.3-39.9; P = .002). In multivariate analysis BL NLR as a discrete variable was independently associated with PSA progression (HR, 3.5; 95% CI, 1.5-8.1; P = .003). NLR at 6 weeks was also associated with duration of benefit; in the favorable NLR category time to PSA progression was 10.8 months, for those who converted to an unfavorable (greater than median) category 4.5 months, and for those remaining in a unfavorable category only 1.5 months (95% CI, 0.5-2.5; P = .003). OS was 33.1 months (95% CI, 24.2-42.0) and 21.9 months (95% CI, 19.3-24.4) for those with an favorable and unfavorable BL NLR, respectively. CONCLUSION Treatment-naive CRPC patients with a high BL or during-treatment NLR appear not to benefit from low-dose corticosteroids. The immunological implications of an unfavorable NLR, and whether corticosteroids might drive prostate cancer progression in patients harboring a high NLR, warrant further study.
Collapse
|
82
|
Evaluation of combination therapy with hydrocortisone, vitamin C, and vitamin E in a rat model of intestine ischemia-reperfusion injury. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s00580-017-2610-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
83
|
Zasada M, Lenart M, Rutkowska-Zapała M, Stec M, Durlak W, Grudzień A, Krzeczkowska A, Mól N, Pilch M, Siedlar M, Kwinta P. Analysis of PD-1 expression in the monocyte subsets from non-septic and septic preterm neonates. PLoS One 2017; 12:e0186819. [PMID: 29049359 PMCID: PMC5648248 DOI: 10.1371/journal.pone.0186819] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
Programmed death-1 (PD-1) receptor system represents a part of recently reported immunoregulatory pathway. PD-1 is an immune checkpoint molecule, which plays an important role in downregulating the immune system proinflammatory activity. Until recently, PD-1 expression was not established on immune cells of the preterm infants. The study objectives were to confirm expression of the PD-1 receptors on the monocytes isolated from very low birth weight newborns (VLBW), and to analyze their expression during the first week of life and late-onset sepsis. Peripheral blood mononuclear cells were isolated from 76 VLBW patients without early-onset sepsis on their 5th day of life (DOL). PD-1 expression was determined on the monocyte subsets (classical, intermediate, non-classical) by flow cytometry. In case of late-onset sepsis (LOS), the same analysis was performed. Our results demonstrated that on the 5th DOL, PD-1 receptors were present in all the monocyte subsets. Children, whose mothers had received antenatal steroids, presented higher absolute numbers of non-classical monocytes with PD-1 expression. Infants born extremely preterm who later developed LOS, initially showed a lower percentage of PD-1 receptor-positive intermediate monocytes in comparison to neonates born very preterm. During LOS, we observed a rise in the percentage of classical monocytes with PD-1 expression. In case of septic shock or fatal outcome, there was a higher percentage and absolute count of intermediate monocytes with PD-1 expression in comparison to children without these complications. In conclusion, monocytes from VLBW children express PD-1 receptors. Antenatal steroid administration seems to induce PD-1 receptor expression in the non-classical monocytes. PD-1 might play a role in immunosuppressive phase of sepsis in the prematurely born children with septic shock and fatal outcome.
Collapse
Affiliation(s)
- Magdalena Zasada
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- * E-mail:
| | - Marzena Lenart
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Magdalena Rutkowska-Zapała
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Małgorzata Stec
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Wojciech Durlak
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Andrzej Grudzień
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Agnieszka Krzeczkowska
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Nina Mól
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Marta Pilch
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Przemko Kwinta
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
84
|
Gautron AS, Juillerat A, Guyot V, Filhol JM, Dessez E, Duclert A, Duchateau P, Poirot L. Fine and Predictable Tuning of TALEN Gene Editing Targeting for Improved T Cell Adoptive Immunotherapy. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 9:312-321. [PMID: 29246309 PMCID: PMC5684446 DOI: 10.1016/j.omtn.2017.10.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 10/11/2017] [Accepted: 10/11/2017] [Indexed: 12/25/2022]
Abstract
Using a TALEN-mediated gene-editing approach, we have previously described a process for the large-scale manufacturing of “off-the-shelf” CAR T cells from third-party donor T cells by disrupting the gene encoding TCRα constant chain (TRAC). Taking advantage of a previously described strategy to control TALEN targeting based on the exclusion capacities of non-conventional RVDs, we have developed highly efficient and specific nucleases targeting a key T cell immune checkpoint, PD-1, to improve engineered CAR T cells’ functionalities. Here, we demonstrate that this approach allows combined TRAC and PDCD1 TALEN processing at the desired locus while eliminating low-frequency off-site processing. Thus, by replacing few RVDs, we provide here an easy and rapid redesign of optimal TALEN combinations. We anticipate that this method can greatly benefit multiplex editing, which is of key importance especially for therapeutic applications where high editing efficiencies need to be associated with maximal specificity and safety.
Collapse
Affiliation(s)
| | | | - Valérie Guyot
- Cellectis SA, 8 Rue de la Croix Jarry, 75013 Paris, France
| | | | - Emilie Dessez
- Cellectis SA, 8 Rue de la Croix Jarry, 75013 Paris, France
| | | | | | - Laurent Poirot
- Cellectis SA, 8 Rue de la Croix Jarry, 75013 Paris, France
| |
Collapse
|
85
|
Overcoming Oncogenic Mediated Tumor Immunity in Prostate Cancer. Int J Mol Sci 2017; 18:ijms18071542. [PMID: 28714919 PMCID: PMC5536030 DOI: 10.3390/ijms18071542] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is being tested intensively in clinical trials for prostate cancer; it includes immune checkpoint inhibition, prostate specific antigen (PSA) vaccines and dendritic cell-based strategies. Despite increasing evidence for clinical responses, the consensus of multiple trials is that prostate cancers are poorly responsive to immunotherapy. Prostate cancer has a high degree of pathological and genetic heterogeneity compared to other cancer types, which may account for immunotherapeutic resistance. This hypothesis also implies that select types of prostate tumors may be differentially responsive to immune-based strategies and that the clinical stage, pathological grade and underlying genetic landscape may be important criteria in identifying tumors that respond to immune therapies. One strategy is to target oncogenic driver pathways in combination with immunotherapies with the goal of overcoming tumor immunity and broadening the number of patients achieving a clinical response. In this analysis, we address the hypothesis that driver oncogenic signaling pathways regulate cancer progression, tumor immunity and resistance to current immune therapeutics in prostate cancer. We propose that increased responsiveness may be achieved through the combined use of immunotherapies and inhibitors targeting tumor cell autonomous pathways that contribute towards anti-tumor immunity in patients with prostate cancer.
Collapse
|
86
|
Vriens BEPJ, Vriens IJH, Aarts MJB, van Gastel SM, van den Berkmortel FWPJ, Smilde TJ, van Warmerdam LJC, van Spronsen DJ, Peer PGM, de Boer M, Tjan-Heijnen VCG. Improved survival for sequentially as opposed to concurrently delivered neoadjuvant chemotherapy in non-metastatic breast cancer. Breast Cancer Res Treat 2017; 165:593-600. [PMID: 28674765 PMCID: PMC5602024 DOI: 10.1007/s10549-017-4364-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/27/2017] [Indexed: 12/22/2022]
Abstract
PURPOSE The INTENS study was designed to determine whether delivering neoadjuvant chemotherapy at a higher dose in a shorter period of time improves outcome of breast cancer patients. METHODS Women with newly diagnosed breast cancer were randomly assigned to neoadjuvant chemotherapy consisting of four cycles of doxorubicin and cyclophosphamide followed by four cycles of docetaxel (AC 60/600-T 100 mg/m2) or six cycles of TAC as triplet chemotherapy (75/50/500 mg/m2) every 3 weeks. The primary outcome was the pathologic complete response (pCR), with disease-free and overall survival as secondary endpoints. RESULTS In total, 201 patients were included. The pCR rates were 28% for patients treated with AC-T and 19% for patients treated with TAC, with an odds ratio of 1.60 (95% CI 0.90-3.21). With a median follow-up of 6 years (range 0.04-8.41 years), the five-year disease-free survival was 81% for patients treated with sequentially AC-T and 71% for patients treated with concurrent triplet TAC chemotherapy with a stratified hazard ratio (HR) of 0.50 (95% CI 0.29-0.86). Five-year overall survival was 84% versus 76%, respectively, with a stratified HR of 0.55 (95% CI 0.29-1.03). CONCLUSIONS No differences were observed between the two treatment arms with respect to pCR rate, but the sequentially delivered chemotherapy outperformed the triplet combination chemotherapy in terms of survival, despite a lower cumulative dose per agent. GOV nr NCT00314977.
Collapse
Affiliation(s)
- B E P J Vriens
- Division of Medical Oncology, Department of Medical Oncology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - I J H Vriens
- Division of Medical Oncology, Department of Medical Oncology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - M J B Aarts
- Division of Medical Oncology, Department of Medical Oncology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - S M van Gastel
- Comprehensive Cancer Centre the Netherlands, Nijmegen, The Netherlands
| | | | - T J Smilde
- Department of Internal Medicine, Jeroen Bosch Hospital, 's Hertogenbosch, The Netherlands
| | - L J C van Warmerdam
- Department of Internal Medicine, Catharina-Hospital, Eindhoven, The Netherlands
| | - D J van Spronsen
- Department of Internal Medicine, Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
| | - P G M Peer
- Biostatistics, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - M de Boer
- Division of Medical Oncology, Department of Medical Oncology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - V C G Tjan-Heijnen
- Division of Medical Oncology, Department of Medical Oncology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | | |
Collapse
|
87
|
Abstract
Glucocorticoid eye drops are one of the most widely used medications in ophthalmology. However, little is known about the effects of glucocorticoids on corneal epithelial cells that are directly exposed to topically-administered glucocorticoids. Here we investigated the effects of prednisolone, a synthetic glucocorticoid analogue frequently used in the clinic, on corneal epithelial cells. Results showed that prednisolone decreased survival of corneal epithelial cells by inhibiting proliferation and inducing apoptosis in a dose-dependent manner. The levels of mitochondrial reactive oxygen species (mtROS), cleaved caspase-3, and -9 were increased by prednisolone. The effects of prednisolone on apoptosis and mtROS were blocked 1) by the glucocorticoid receptor (GR) antagonist RU-38486, 2) in cells with GR siRNA knockdown, and 3) by treatment with N-acetylcysteine. Transcript levels of pro-inflammatory cytokines were increased in corneal epithelial cells upon hyperosmolar stress, but repressed by prednisolone. In NOD.B10.H2b mice, topical administration of 1% prednisolone increased apoptotic cells in the corneal epithelium. Together, data indicate that prednisolone induces apoptosis in corneal epithelial cells through GR and the intrinsic pathway involving mtROS, caspase-9, and -3. The pro-apoptotic effects of glucocorticoids along with their anti-inflammatory effects should be considered when glucocorticoid eye drops are used in patients with ocular surface disease.
Collapse
|
88
|
Li JH, Pang NN, Zhang ZH, Zhang R, Chen G, Qu JH. [PD-1/PD-L1 expression and its implications in patients with chronic lymphocytic leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2017; 38:198-203. [PMID: 28395442 PMCID: PMC7348379 DOI: 10.3760/cma.j.issn.0253-2727.2017.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Indexed: 12/26/2022]
Abstract
Objective: To observe the expression levels of PD-1/PD-L1 costimulatory molecules and explore the clinical significance in patients with chronic lymphocytic leukemia (CLL) . Methods: The expression of PD-1/PD-L1 in peripheral blood CD8(+) T cells, CD4(+)T cells, CD19(+)B, and dendrites cells (DC) was detected by flow cytometry in 57 CLL patients and 20 healthy controls. The correlations of PD-1/PD-L1 expression with disease stage, CD38 expression, ZAP-70 expression, chromosome karyotype abnormality and β(2)-MG expression were analyzed. Results: ①Compared with control, CLL patients, including 39 males and 18 females with the median age of (63.7±10.7) years, had no statistically significant difference in age and gender (P>0.05) . CLL patients had the higher PD-1/PD-L1 expression than healthy controls (P<0.05) . ②In Rai staging, the later the stage, the higher expression of PD-1/PD-L1 (P<0.05) . ③PD-1 expression in CD8(+)CD38(+) group (11 cases) was higher than that in CD8(+)CD38(-) group (46 cases) (P=0.004) , and CD8(+) poor prognosis chromosome group (14 cases) also had significant higher PD-1 expression than CD8(+)good prognosis chromosome group (28 cases) (P=0.004) . ④The expression of PD-L1 was higher in CD38(+)group, ZAP-70(+)group, and poor prognosis group, as compared to that in CD38(-)group (P=0.002) , in ZAP-70(-)group (P<0.001) , in good prognosis group (P=0.023) . There was no correlation between the expression of PD-1/PD-L1 and β(2)-MG (P>0.05) . Conclusion: This data reveals that PD-1/PD-L1 was highly expressed in CLL patients. Its expression levels were correlated with Rai stage, CD38, ZAP-70, chromosome karyotype, but not with β(2)-MG. PD-1/PD-L1 may be a prognostic factor in patients with CLL.
Collapse
Affiliation(s)
- J H Li
- Xinjiang Medical University First Affiliated Hospital Blood Disease Center, Xinjiang Uygur Autonomous Region of Hematology, Urumqi 830054, China
| | | | | | | | | | | |
Collapse
|
89
|
Grodzinsky AJ, Wang Y, Kakar S, Vrahas MS, Evans CH. Intra-articular dexamethasone to inhibit the development of post-traumatic osteoarthritis. J Orthop Res 2017; 35:406-411. [PMID: 27176565 PMCID: PMC5604325 DOI: 10.1002/jor.23295] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 05/06/2016] [Indexed: 02/04/2023]
Abstract
UNLABELLED Injury to the joint provokes a number of local pathophysiological changes, including synthesis of inflammatory cytokines, death of chondrocytes, breakdown of the extra-cellular matrix of cartilage, and reduced synthesis of matrix macromolecules. These processes combine to engender the subsequent development of post-traumatic osteoarthritis (PTOA). To prevent this from happening, it is necessary to inhibit these disparate responses to injury; given their heterogeneity, this is challenging. However, dexamethasone has the necessary pleiotropic properties required of a drug for this purpose. Using in vitro models, we have shown that low doses of dexamethasone sustain the synthesis of cartilage proteoglycans while inhibiting their breakdown after injurious compression in the presence or absence of inflammatory cytokines. Under these conditions, dexamethasone is non-toxic and maintains the viability of chondrocytes exposed chronically to such cytokines as interleukin (IL) -1, IL-6, and tumor necrosis factor-α. Moreover, the anti-inflammatory properties of dexamethasone have been appreciated for decades. In view of this information, we have initiated a pilot clinical study to determine whether a single, intra-articular injection of dexamethasone into the wrist shows promise in preventing PTOA after intra-articular fracture of the distal radius. CLINICAL SIGNIFICANCE Suppressing the various etiopathophysiological responses to injury in the joint is an attractive strategy for lowering the clinical burden of PTOA. The intra-articular administration of dexamethasone soon after injury offers a simple and inexpensive means of accomplishing this. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:406-411, 2017.
Collapse
Affiliation(s)
- Alan J. Grodzinsky
- Departments of Biological, Electrical and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Yang Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Sanjeev Kakar
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, MN
| | - Mark S. Vrahas
- Department of Orthopaedic Surgery, Harvard Medical School
| | | |
Collapse
|
90
|
Aarslev K, Dige A, Greisen SR, Kreutzfeldt M, Jessen N, Vilstrup H, Deleuran B, Grønbæk H. Soluble programmed death-1 levels are associated with disease activity and treatment response in patients with autoimmune hepatitis. Scand J Gastroenterol 2017; 52:93-99. [PMID: 27604386 DOI: 10.1080/00365521.2016.1233576] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE Autoimmune hepatitis (AIH) is a chronic liver disease caused by impaired immune regulation. Programmed death-1 (PD-1) is an inhibitory receptor mainly expressed by T cells and with its ligands, PD-L1 and PD-L2 present on antigen-presenting cells. We hypothesised the PD-1 axis to be impaired in AIH and investigated systemic levels of soluble(s) PD-1 and T cells ability to up-regulate PD-1 following in vitro activation in AIH patients. MATERIALS AND METHODS We included 67 AIH patients; 9 with active disease, 31 responders and 27 incomplete-responders to standard therapy. Forty-seven healthy controls (HC) were included for comparison. Soluble PD-1 was measured by enzyme-linked immunosorbent assay. The PD-1 expression on T cells was measured using flow cytometry before and after 48-h stimulation in vitro with CD3/CD28 in 13 AIH patients and 10 HC. RESULTS Soluble PD-1 was significantly elevated in AIH patients with active disease [0.24 ng/mL (range 0.16-0.28)] and in incomplete responders to standard therapy [0.17 (0.11-0.22)] compared with responders [0.11 (0.08-0.16), p = .008 and p = .01, respectively] and HC [0.12 (0.05-0.16), p = .02, both]. Following in vitro activation, PD-1 was significantly up-regulated (3.3-fold) on CD4+ T cells from AIH patients compared with HC (1.5-fold) (p = .0006). CONCLUSIONS AIH patients with active disease and incomplete response to standard treatment have similarly increased sPD-1 levels. Further, AIH patients have increased ability to up-regulate PD-1 following in vitro activation. Together these data suggests an impaired PD-1 axis in AIH.
Collapse
Affiliation(s)
- Kristian Aarslev
- a Department of Hepatology and Gastroenterology , Aarhus University Hospital , Aarhus C , Denmark
| | - Anders Dige
- a Department of Hepatology and Gastroenterology , Aarhus University Hospital , Aarhus C , Denmark
| | - Stinne R Greisen
- b Department of Biomedicine , Aarhus University , Aarhus C , Denmark
| | - Martin Kreutzfeldt
- a Department of Hepatology and Gastroenterology , Aarhus University Hospital , Aarhus C , Denmark
| | - Niels Jessen
- c Department of Endocrinology (MEA) , Aarhus University Hospital , Aarhus C , Denmark
| | - Hendrik Vilstrup
- a Department of Hepatology and Gastroenterology , Aarhus University Hospital , Aarhus C , Denmark
| | - Bent Deleuran
- b Department of Biomedicine , Aarhus University , Aarhus C , Denmark.,d Department of Rheumatology , Aarhus University Hospital , Aarhus C , Denmark
| | - Henning Grønbæk
- a Department of Hepatology and Gastroenterology , Aarhus University Hospital , Aarhus C , Denmark
| |
Collapse
|
91
|
Azher S, Azami O, Amato C, McCullough M, Celentano A, Cirillo N. The Non-Conventional Effects of Glucocorticoids in Cancer. J Cell Physiol 2016; 231:2368-73. [PMID: 27115293 DOI: 10.1002/jcp.25408] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 04/25/2016] [Indexed: 12/26/2022]
Abstract
Synthetic corticosteroids are widely used for the treatment of a variety of diseases, including pre-malignant and malignant conditions. In striking contrast, recent evidence suggests that corticosteroids can bear tumor-promoting effects in solid tumors of epithelial origin. We have recently shown that epithelial tissues, including the mucosa of the oral cavity and the skin, are able to modulate the local concentration of active corticosteroids and to produce steroids de novo. This has important clinical and physiopathological implications, because tissue-specific regulation of glucocorticoids plays a key role in the overall effect of these molecules. In the present review of the current English literature, performed using MEDLINE/PubMed/Ovid databases, we collected published evidence to demonstrate that corticosteroids induce effects that are more complex and controversial than previously acknowledged. Published studies clearly demonstrate that this class of molecules influences pathophysiological processes that are strictly related to malignancy, providing the rationale for further investigation. J. Cell. Physiol. 231: 2368-2373, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Simra Azher
- Melbourne Dental School, University of Melbourne, Carlton, Victoria, Australia
| | - Omid Azami
- Melbourne Dental School, University of Melbourne, Carlton, Victoria, Australia
| | - Caterina Amato
- Melbourne Dental School, University of Melbourne, Carlton, Victoria, Australia
| | - Michael McCullough
- Melbourne Dental School, University of Melbourne, Carlton, Victoria, Australia
- Oral Health Cooperative Research Centre (CRC), University of Melbourne, Carlton, Victoria, Australia
| | - Antonio Celentano
- Melbourne Dental School, University of Melbourne, Carlton, Victoria, Australia
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University Federico II of Naples, Naples, Italy
| | - Nicola Cirillo
- Melbourne Dental School, University of Melbourne, Carlton, Victoria, Australia
- Oral Health Cooperative Research Centre (CRC), University of Melbourne, Carlton, Victoria, Australia
| |
Collapse
|
92
|
Adrenal-Derived Hormones Differentially Modulate Intestinal Immunity in Experimental Colitis. Mediators Inflamm 2016; 2016:4936370. [PMID: 27403034 PMCID: PMC4923585 DOI: 10.1155/2016/4936370] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 05/19/2016] [Indexed: 01/09/2023] Open
Abstract
The adrenal glands are able to modulate immune responses through neuroimmunoendocrine interactions and cortisol secretion that could suppress exacerbated inflammation such as in inflammatory bowel disease (IBD). Therefore, here we evaluated the role of these glands in experimental colitis induced by 3% dextran sulfate sodium (DSS) in C57BL/6 mice subjected to adrenalectomy, with or without glucocorticoid (GC) replacement. Mice succumbed to colitis without adrenals with a higher clinical score and augmented systemic levels of IL-6 and lower LPS. Furthermore, adrenalectomy negatively modulated systemic regulatory markers. The absence of adrenals resulted in augmented tolerogenic lamina propria dendritic cells but no compensatory local production of corticosterone and decreased mucosal inflammation associated with increased IFN-γ and FasL in the intestine. To clarify the importance of GC in this scenario, GC replacement in adrenalectomized mice restored different markers to the same degree of that observed in DSS group. Finally, this is the first time that adrenal-derived hormones, especially GC, were associated with the differential local modulation of the gut infiltrate, also pointing to a relationship between adrenalectomy and the modulation of systemic regulatory markers. These findings may elucidate some neuroimmunoendocrine mechanisms that dictate colitis outcome.
Collapse
|
93
|
Dexamethasone Suppresses Oxysterol-Induced Differentiation of Monocytic Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2915382. [PMID: 27340507 PMCID: PMC4906206 DOI: 10.1155/2016/2915382] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/14/2016] [Accepted: 05/15/2016] [Indexed: 11/21/2022]
Abstract
Oxysterol like 27-hydroxycholesterol (27OHChol) has been reported to induce differentiation of monocytic cells into a mature dendritic cell phenotype. We examined whether dexamethasone (Dx) affects 27OHChol-induced differentiation using THP-1 cells. Treatment of monocytic cells with Dx resulted in almost complete inhibition of transcription and surface expression of CD80, CD83, and CD88 induced by 27OHChol. Elevated surface levels of MHC class I and II molecules induced by 27OHChol were reduced to basal levels by treatment with Dx. A decreased endocytosis ability caused by 27OHChol was recovered by Dx. We also examined effects of Dx on expression of CD molecules involved in atherosclerosis. Increased levels of surface protein and transcription of CD105, CD137, and CD166 by treatment with 27OHChol were significantly inhibited by cotreatment with Dx. These results indicate that Dx inhibits 27OHChol-induced differentiation of monocytic cells into a mature dendritic cell phenotype and expression of CD molecules whose levels are associated with atherosclerosis. In addition, we examined phosphorylation of AKT induced by 27OHChol and effect of Dx, where cotreatment with Dx inhibited the phosphorylation of AKT. The current study reports that Dx regulates oxysterol-mediated dendritic cell differentiation of monocytic cells.
Collapse
|