1
|
Li Y, Sun L, Zhou Q, Lee AJ, Wang L, Zhang R, Wang S. Effects of opioid drugs on immune function in cancer patients. Biomed Pharmacother 2024; 175:116665. [PMID: 38701564 DOI: 10.1016/j.biopha.2024.116665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/05/2024] Open
Abstract
Opioid receptor agonists are often used when cancer patients undergo surgery or analgesic treatment. As analgesics in clinical care, opioids can provide intraoperative or to chronic cancer pain relief. Immune function plays an important role in anti-cancer therapy, with cellular immunity, comprised principally of T-lymphocytes and natural killer cells, representing the primary anti-cancer immune response. However, it remains unclear whether immune function is further affected with the use of opioids in already immunocompromised cancer patients. This article provides a review of the effects of commonly used clinical opioids, including morphine, oxycodone, fentanyl and tramadol, on immune function in cancer patients. It provides a summary of current evidence regarding the immunomodulatory effects of opioids in the cancer setting and mechanisms underlying these interactions.
Collapse
Affiliation(s)
- Yunqi Li
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Lina Sun
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China.
| | - Qinglian Zhou
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - An Jie Lee
- Biological Sciences, University of California San Diego, La Jolla, CA 92093, United States
| | - Lingyan Wang
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Rui Zhang
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China.
| | - Shoushi Wang
- Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao 266042, China.
| |
Collapse
|
2
|
Lee JE, Jung H, Byun SH, Park JM, Yeo J, Jeon Y, Lee SW, Park SS, Lim DG, Kim SO, Kwak KH. Effect of Dexmedetomidine Preconditioning on Hepatic Ischemia-Reperfusion Injury in Acute Hyperglycemic Rats. Transplant Proc 2023; 55:2478-2486. [PMID: 37867004 DOI: 10.1016/j.transproceed.2023.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/08/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND Acute hyperglycemia frequently occurs in stressful situations, including liver transplantation or hepatic surgery, which may affect the protective effects of dexmedetomidine preconditioning and increase postoperative mortality. Therefore, this study aimed to investigate the effects of dexmedetomidine on hepatic ischemia-reperfusion injury in acute hyperglycemia. METHODS Thirty-six Sprague-Dawley rats were randomly assigned to 6 groups, including a combination between 2 glycemic (normo- and hyperglycemia) and 3 ischemia-reperfusion conditions (sham, ischemia-reperfusion only, and dexmedetomidine plus ischemia-reperfusion). Dexmedetomidine 70 μg/kg was preconditioned 30 minutes before ischemic injury. After 6 hours of reperfusion, serum aminotransferase levels were measured to confirm the hepatic tissue injury. Furthermore, inflammatory (nuclear factor-κb, tumor necrosis factor-α, and interleukin-6) and oxidative stress markers (malondialdehyde and superoxide dismutase) were detected. RESULTS Ischemia-reperfusion injury significantly increased the serum levels of aminotransferase and inflammatory and oxidative stress markers. These ischemia-reperfusion-induced changes were further exacerbated in hyperglycemia and were significantly attenuated by dexmedetomidine preconditioning. However, the effects of dexmedetomidine in hyperglycemia were lesser than those in normoglycemia (P < .05 for aminotransferases, inflammatory markers, malondialdehyde, and superoxide dismutase). CONCLUSIONS These findings suggest that the protective effects of dexmedetomidine preconditioning may be intact against hepatic ischemia-reperfusion injury in acute hyperglycemia. Although its effects appeared to be relatively reduced, this may be because of the increase in oxidative stress and inflammatory response caused by acute hyperglycemia. To determine whether the effects of dexmedetomidine itself would be impaired in hyperglycemia, further study is needed.
Collapse
Affiliation(s)
- Jeong Eun Lee
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Hoon Jung
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Sung-Hye Byun
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Jun-Mo Park
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Jinseok Yeo
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Younghoon Jeon
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - See Woo Lee
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Sung-Sik Park
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Dong Gun Lim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Si-Oh Kim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Kyung-Hwa Kwak
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea.
| |
Collapse
|
3
|
Wang Q, Liu Z, Tang S, Wu Z. Morphine suppresses the immune function of lung cancer by up-regulating MAEL expression. BMC Pharmacol Toxicol 2022; 23:92. [PMID: 36476246 PMCID: PMC9730686 DOI: 10.1186/s40360-022-00632-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Patients with cancer rely on morphine for analgesia, while studies have indicated morphine can induce immunosuppression in cancer. Therefore, investigating the immunosuppressive roles and molecular mechanism of morphine on lung cancer progression is imperative. METHODS Lactate dehydrogenase (LDH) release assay was used to determine the cytotoxicity of morphine to lung cancer cells. The percentage of CD4+ and CD8+ T cells was detected by flow cytometry. In addition, Maelstrom (MAEL), Nrf2, and PTEN were determined by western blot and RT-qPCR. Immune factors programmed death-ligand 1 (PD-L1), transforming growth factor (TGF-β), interleukin (IL)-10, and IL-2 were determined by western blot and ELISA assay. RESULTS Morphine increased the levels of PD-L1, TGF-β, and IL-10, while decreased IL-2 level. Morphine enhanced MAEL expression in A549 cells and H460 cells. Morphine up-regulated Nrf2 and down-regulated PTEN, and morphine-induced MAEL up-regulation was reversed by PTEN. However, MAEL silencing inhibited the enhanced effects of morphine on cell viability and proliferation of A549 cells. Furthermore, morphine treatment reduced the LDH release and the percentage of CD8+ T cells, and increased the ratio of CD4+/CD8+ T cells and tumor weight. Meanwhile, MAEL silencing reversed the effects of morphine on immune factors (PD-L1, TGF-β, IL-10, and IL-2), the percentage of CD8+ T cells, and the ratio of CD4+/CD8+ T cells. CONCLUSION Morphine activated MAEL in lung cancer cells by Nrf2/PTEN pathway and regulated the immune factors, thereby promoting tumor immune escape.
Collapse
Affiliation(s)
- Qichao Wang
- grid.411634.50000 0004 0632 4559Department of Oncology II, Dalian Fifth People’s Hospital, No. 890, Huanghe Road, Shahekou District, Dalian City, 116021 Liaoning Province China
| | - Zhenfu Liu
- Department of Anesthesiology, Zaozhuang Hospital of Zaozhuang Mining Group, No. 188, Shengli Road, Zaozhuang City, 277100 Shandong Province China
| | - Shuhong Tang
- grid.411634.50000 0004 0632 4559Department of Oncology II, Dalian Fifth People’s Hospital, No. 890, Huanghe Road, Shahekou District, Dalian City, 116021 Liaoning Province China
| | - Zhen Wu
- grid.452582.cDepartment of Anesthesiology, The Fourth Hospital of Hebei Medical University, No.12, Jiankang Road, Shijiazhuang City, 050000 Hebei Province China
| |
Collapse
|
4
|
Targeted nanomedicines remodeling immunosuppressive tumor microenvironment for enhanced cancer immunotherapy. Acta Pharm Sin B 2022; 12:4327-4347. [PMID: 36561994 PMCID: PMC9764075 DOI: 10.1016/j.apsb.2022.11.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/03/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Cancer immunotherapy has significantly flourished and revolutionized the limited conventional tumor therapies, on account of its good safety and long-term memory ability. Discouragingly, low patient response rates and potential immune-related side effects make it rather challenging to literally bring immunotherapy from bench to bedside. However, it has become evident that, although the immunosuppressive tumor microenvironment (TME) plays a pivotal role in facilitating tumor progression and metastasis, it also provides various potential targets for remodeling the immunosuppressive TME, which can consequently bolster the effectiveness of antitumor response and tumor suppression. Additionally, the particular characteristics of TME, in turn, can be exploited as avenues for designing diverse precise targeting nanomedicines. In general, it is of urgent necessity to deliver nanomedicines for remodeling the immunosuppressive TME, thus improving the therapeutic outcomes and clinical translation prospects of immunotherapy. Herein, we will illustrate several formation mechanisms of immunosuppressive TME. More importantly, a variety of strategies concerning remodeling immunosuppressive TME and strengthening patients' immune systems, will be reviewed. Ultimately, we will discuss the existing obstacles and future perspectives in the development of antitumor immunotherapy. Hopefully, the thriving bloom of immunotherapy will bring vibrancy to further exploration of comprehensive cancer treatment.
Collapse
|
5
|
Yu Y, Liu Y, Yao L, Shen Y, Sun Q, Sha W. Factors Influencing False-Negative Results of QuantiFERON-TB Gold In-Tube (QFT-GIT) in Active Tuberculosis and the Desirability of Resetting Cutoffs for Different Populations: A Retrospective Study. Trop Med Infect Dis 2022; 7:tropicalmed7100278. [PMID: 36288019 PMCID: PMC9607193 DOI: 10.3390/tropicalmed7100278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 11/07/2022] Open
Abstract
Objectives The value of QuantiFERON-TB Gold In-Tube (QFT-GIT) in the diagnosis of TB varies by population, comorbidities, and other factors. In this study, we aimed to investigate factors that influence false-negative results of QFT-GIT test in the diagnosis of TB as well as the impact of different cutoffs on the diagnostic value. Methods A total of 3562 patients who underwent QFT-GIT tests at Shanghai Pulmonary Hospital were enrolled retrospectively between May 2016 and May 2017. False-negative and false-positive results were analyzed using different clinical stratifications. The optimal cutoff values were established under different clinical conditions. Results Positive QFT-GIT results greatly shortened the time taken to diagnose smear-negative TB. The factors of age, smear and culture results, site of TB, comorbidity with tumors, white blood cell count, neutrophil count, and CD4/CD8 ratio were significantly correlated with false-negative QFT-GIT results (p < 0.05). Personalized cutoff values were established according to different influencing factors. The results showed high consistency between the smear-negative and total populations. Conclusion QFT-GIT can facilitate the early diagnosis of smear-negative TB. The diagnostic performance of the QFT-GIT test in the diagnosis of active TB was shown to be affected by many clinical factors. Personalized cutoff values may have superior value in the identification of active tuberculosis under different conditions.
Collapse
Affiliation(s)
- Yuanyuan Yu
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Clinical and Research Center for Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Yidian Liu
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Clinical and Research Center for Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Lan Yao
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Clinical and Research Center for Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Yanheng Shen
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Clinical and Research Center for Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Qin Sun
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Clinical and Research Center for Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Correspondence: (Q.S.); (W.S.)
| | - Wei Sha
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Clinical and Research Center for Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Correspondence: (Q.S.); (W.S.)
| |
Collapse
|
6
|
Di Pompo G, Cortini M, Baldini N, Avnet S. Acid Microenvironment in Bone Sarcomas. Cancers (Basel) 2021; 13:cancers13153848. [PMID: 34359749 PMCID: PMC8345667 DOI: 10.3390/cancers13153848] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/24/2021] [Accepted: 07/28/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Although rare, malignant bone sarcomas have devastating clinical implications for the health and survival of young adults and children. To date, efforts to identify the molecular drivers and targets have focused on cancer cells or on the interplay between cancer cells and stromal cells in the tumour microenvironment. On the contrary, in the current literature, the role of the chemical-physical conditions of the tumour microenvironment that may be implicated in sarcoma aggressiveness and progression are poorly reported and discussed. Among these, extracellular acidosis is a well-recognized hallmark of bone sarcomas and promotes cancer growth and dissemination but data presented on this topic are fragmented. Hence, we intended to provide a general and comprehensive overview of the causes and implications of acidosis in bone sarcoma. Abstract In bone sarcomas, extracellular proton accumulation is an intrinsic driver of malignancy. Extracellular acidosis increases stemness, invasion, angiogenesis, metastasis, and resistance to therapy of cancer cells. It reprograms tumour-associated stroma into a protumour phenotype through the release of inflammatory cytokines. It affects bone homeostasis, as extracellular proton accumulation is perceived by acid-sensing ion channels located at the cell membrane of normal bone cells. In bone, acidosis results from the altered glycolytic metabolism of bone cancer cells and the resorption activity of tumour-induced osteoclasts that share the same ecosystem. Proton extrusion activity is mediated by extruders and transporters located at the cell membrane of normal and transformed cells, including vacuolar ATPase and carbonic anhydrase IX, or by the release of highly acidic lysosomes by exocytosis. To date, a number of investigations have focused on the effects of acidosis and its inhibition in bone sarcomas, including studies evaluating the use of photodynamic therapy. In this review, we will discuss the current status of all findings on extracellular acidosis in bone sarcomas, with a specific focus on the characteristics of the bone microenvironment and the acid-targeting therapeutic approaches that are currently being evaluated.
Collapse
Affiliation(s)
- Gemma Di Pompo
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
| | - Margherita Cortini
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
| | - Nicola Baldini
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
- Correspondence:
| |
Collapse
|
7
|
Xie H, Shu C, Bai H, Sun P, Liu H, Qi J, Li S, Ye C, Gao F, Yuan M, Chen Y, Pan M, Yang X, Ma Y. A therapeutic HPV16 E7 vaccine in combination with active anti-FGF-2 immunization synergistically elicits robust antitumor immunity in mice. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102254. [PMID: 32615335 DOI: 10.1016/j.nano.2020.102254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 05/28/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022]
Abstract
FGF-2 accumulates in many tumor tissues and is closely related to the development of tumor angiogenesis and the immunosuppressive microenvironment. This study aimed to investigate whether active immunization against FGF-2 could modify antitumor immunity and enhance the efficacy of an HPV16 E7-specific therapeutic vaccine. Combined immunization targeting both FGF-2 and E7 significantly suppressed tumor growth, which was accompanied by significantly increased levels of IFN-γ-expressing splenocytes and effector CD8 T cells and decreased levels of immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells(MDSCs) in both the spleen and tumor; in addition, the levels of FGF-2 and neovascularization in tumors were decreased in the mice receiving the combined immunization, and tumor cell apoptosis was promoted. The combination of an HPV16 E7-specific vaccine and active immunization against FGF-2 significantly enhances antitumor immune responses in mice with TC-1 tumors, indicating a promising strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- Hanghang Xie
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease
| | - Congyan Shu
- Sichuan Institute for Food and Drug Control, Chengdu, China
| | - Hongmei Bai
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease
| | - Pengyan Sun
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Center for Disease Control and Prevention; Kunming, China
| | - Hongxian Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease
| | - Jialong Qi
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease
| | - Sijin Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease
| | - Chao Ye
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease
| | - Fulan Gao
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease
| | - Mingcui Yuan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease
| | - Yongjun Chen
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease
| | - Manchang Pan
- Department of Burn, The Second Affiliated Hospital, Kunming Medical University,Kunming, China
| | - Xu Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China; Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease.
| |
Collapse
|
8
|
Wang K, Wu M, Xu J, Wu C, Zhang B, Wang G, Ma D. Effects of dexmedetomidine on perioperative stress, inflammation, and immune function: systematic review and meta-analysis. Br J Anaesth 2019; 123:777-794. [DOI: 10.1016/j.bja.2019.07.027] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/26/2019] [Accepted: 07/03/2019] [Indexed: 01/06/2023] Open
|
9
|
Singh A, Koutsoumpli G, van de Wall S, Daemen T. An alphavirus-based therapeutic cancer vaccine: from design to clinical trial. Cancer Immunol Immunother 2019; 68:849-859. [PMID: 30465060 PMCID: PMC11028389 DOI: 10.1007/s00262-018-2276-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/09/2018] [Indexed: 01/25/2023]
Abstract
Cancer immunotherapy has greatly advanced in recent years. Most immunotherapeutic strategies are based on the use of immune checkpoint blockade to unleash antitumor immune responses or on the induction or adoptive transfer of immune effector cells. We aim to develop therapeutic vaccines based on recombinant Semliki Forest virus vectors to induce tumor-specific effector immune cells. In this review, we describe our ongoing work on SFV-based vaccines targeted against human papillomavirus- and hepatitis C virus-related infections and malignancies, focusing on design, delivery, combination strategies, preclinical efficacy and product development for a first-in-man clinical trial with an HPV-specific vaccine.
Collapse
Affiliation(s)
- Amrita Singh
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, HPC EB88, PO Box 30.001, 9700RB, Groningen, The Netherlands
| | - Georgia Koutsoumpli
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, HPC EB88, PO Box 30.001, 9700RB, Groningen, The Netherlands
| | - Stephanie van de Wall
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, HPC EB88, PO Box 30.001, 9700RB, Groningen, The Netherlands
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Toos Daemen
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, HPC EB88, PO Box 30.001, 9700RB, Groningen, The Netherlands.
| |
Collapse
|
10
|
Hou M, Zhou N, Li H, Wang B, Wang X, Wang X, Jiang T, Wang K, Xue F. Morphine and ketamine treatment suppress the differentiation of T helper cells of patients with colorectal cancer in vitro. Exp Ther Med 2019; 17:935-942. [PMID: 30651883 DOI: 10.3892/etm.2018.7035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 11/11/2016] [Indexed: 11/06/2022] Open
Abstract
There have been conflicting reports regarding the effects of anesthetic and analgesic drugs on immune function in patients with cancer. The aim of the present study was to investigate changes to T helper (Th) cell populations in patients with colorectal cancer (CRC) and to assess the effects of morphine and ketamine on the differentiation of Th cells harvested from patients with CRC in vitro. Peripheral blood samples were extracted from 20 patients with CRC and 20 healthy participants. Peripheral blood mononuclear cells were isolated and incubated in a solution containing phorbol-myristate-acetate (PMA) and ionomycin in the presence or absence of morphine or various ketamine concentrations (25, 50, and 100 µM). Samples were analyzed 4 h later. Th1 and Th2 cells were significantly increased by PMA and ionomycin stimulation; however, Th1 cells and the Th1/Th2 ratio in PMA and ionomycin treatments were significantly decreased in the CRC group compared with the control group. Following incubation with PMA and ionomycin, morphine significantly decreased Th1 cells and the Th1/Th2 ratio in the CRC group. Ketamine did not significantly affect levels of Th1 or Th2 cells or the Th1/Th2 ratio at a concentration of 25 µM; however, a significant increase in the Th1/Th2 ratio was observed at a concentration of 50 µM and, at 100 µM, a significant decrease in Th1 and Th2 cells and an increase in the Th1/Th2 ratio were observed. The present study suggests that CRC may shift the balance of Th1/Th2 towards Th2 by inducing an immunological response, morphine is able to suppress the differentiation of Th cells and decreases the Th1/Th2 ratio, and ketamine may affect the differentiation of Th cells in a dose-dependent manner.
Collapse
Affiliation(s)
- Min Hou
- Department of Anesthesiology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China.,Department of Anesthesiology, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Naibao Zhou
- Department of Anesthesiology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Hao Li
- Department of Anesthesiology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Baosheng Wang
- Department of Anesthesiology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Xiuqin Wang
- Department of Anesthesiology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Xingwu Wang
- Department of Anesthesiology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Tao Jiang
- Department of Anesthesiology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Kaiguo Wang
- Department of Anesthesiology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Fushan Xue
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
11
|
Yuan Z, Yang H, Wei Y. Combined induction with anti-PD-1 and anti-CTLA-4 antibodies provides synergistic antitumor effects in DC-CIK cells in renal carcinoma cell lines. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:123-132. [PMID: 31933726 PMCID: PMC6944006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 11/13/2018] [Indexed: 06/10/2023]
Abstract
Immune escape of cancer cells has become the main challenge in the immunocytotherapy field. In this study, we analyzed the cytotoxicity of DC-CIK cells induced by anti-PD-1 and anti-CTLA-4 antibodies in RCC cell lines. Flow cytometry analysis was performed to analyze the immune phenotypes of DC-CIK cells. Click-iT EdU assay was performed to analyze the proliferation of DC-CIK cells. ELISA analysis was performed to detect the expression of cytokines in DC-CIK cells. Compared with DC-CIK cells without any treatment, the growth inhibition rate was significantly higher in the other three groups. Moreover, combined induction with anti-PD-1 plus anti-CTLA-4 antibodies provides synergistic antitumor effects of DC-CIK cells in renal carcinoma cell lines. The combined treatment promoted DC-CIK cell proliferation and differentiation into CD3+CD56+ NKT cells and CD3+CD8+ CTL cells. Compared with the control group, combined treatment significantly up-regulated the secretion of immune-stimulatory cytokines, such as IFN-γ and TNF-α, and down-regulated the secretion of the immunosuppressive cytokine IL-10. Furthermore, the co-induction promoted the early activation of DC-CIK cells. These results indicated the co-induction with anti-PD-1 plus anti-CTLA-4 antibodies improved antitumor effects of DC-CIK cells by promoting proliferation, differentiation, and early activation and regulating the secretion of immune-stimulatory and suppressive cytokines in renal carcinoma cell lines.
Collapse
Affiliation(s)
- Zhaohu Yuan
- Department of Blood Transfusion, Guangzhou First People’s Hospital, School of Medicine, South China University of TechnologyGuangzhou 510180, Guangdong, China
- Guangdong Engineering Research Center of Precise TransfusionGuangzhou 510180, Guangdong, China
| | - Huikuan Yang
- Department of Blood Transfusion, Guangzhou First People’s Hospital, School of Medicine, South China University of TechnologyGuangzhou 510180, Guangdong, China
- Guangdong Engineering Research Center of Precise TransfusionGuangzhou 510180, Guangdong, China
| | - Yaming Wei
- Department of Blood Transfusion, Guangzhou First People’s Hospital, School of Medicine, South China University of TechnologyGuangzhou 510180, Guangdong, China
- Guangdong Engineering Research Center of Precise TransfusionGuangzhou 510180, Guangdong, China
| |
Collapse
|
12
|
Damgaci S, Ibrahim‐Hashim A, Enriquez‐Navas PM, Pilon‐Thomas S, Guvenis A, Gillies RJ. Hypoxia and acidosis: immune suppressors and therapeutic targets. Immunology 2018; 154:354-362. [PMID: 29485185 PMCID: PMC6002221 DOI: 10.1111/imm.12917] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 02/08/2018] [Accepted: 02/13/2018] [Indexed: 12/12/2022] Open
Abstract
Due to imbalances between vascularity and cellular growth patterns, the tumour microenvironment harbours multiple metabolic stressors including hypoxia and acidosis, which have significant influences on remodelling both tumour and peritumoral tissues. These stressors are also immunosuppressive and can contribute to escape from immune surveillance. Understanding these effects and characterizing the pathways involved can identify new targets for therapy and may redefine our understanding of traditional anti-tumour therapies. In this review, the effects of hypoxia and acidosis on tumour immunity will be summarized, and how modulating these parameters and their sequelae can be a useful tool for future therapeutic interventions is discussed.
Collapse
Affiliation(s)
- Sultan Damgaci
- Department of Cancer PhysiologyH. Lee Moffitt Cancer CenterTampaFLUSA
- Institute of Biomedical EngineeringBogazici UniversityIstanbulTurkey
| | | | | | - Shari Pilon‐Thomas
- Department of ImmunologyH. Lee Moffitt Cancer CenterTampaFLUSA
- Department of Cutaneous OncologyH. Lee Moffitt Cancer CenterTampaFLUSA
| | - Albert Guvenis
- Institute of Biomedical EngineeringBogazici UniversityIstanbulTurkey
| | - Robert J. Gillies
- Department of Cancer PhysiologyH. Lee Moffitt Cancer CenterTampaFLUSA
- Department of RadiologyH. Lee Moffitt Cancer CenterTampaFLUSA
| |
Collapse
|
13
|
Berrong Z, Mkrtichyan M, Ahmad S, Webb M, Mohamed E, Okoev G, Matevosyan A, Shrimali R, Abu Eid R, Hammond S, Janik JE, Khleif SN. Antigen-Specific Antitumor Responses Induced by OX40 Agonist Are Enhanced by the IDO Inhibitor Indoximod. Cancer Immunol Res 2018; 6:201-208. [PMID: 29305519 DOI: 10.1158/2326-6066.cir-17-0223] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/26/2017] [Accepted: 12/20/2017] [Indexed: 11/16/2022]
Abstract
Although an immune response to tumors may be generated using vaccines, so far, this approach has only shown minimal clinical success. This is attributed to the tendency of cancer to escape immune surveillance via multiple immune suppressive mechanisms. Successful cancer immunotherapy requires targeting these inhibitory mechanisms along with enhancement of antigen-specific immune responses to promote sustained tumor-specific immunity. Here, we evaluated the effect of indoximod, an inhibitor of the immunosuppressive indoleamine-(2,3)-dioxygenase (IDO) pathway, on antitumor efficacy of anti-OX40 agonist in the context of vaccine in the IDO- TC-1 tumor model. We demonstrate that although the addition of anti-OX40 to the vaccine moderately enhances therapeutic efficacy, incorporation of indoximod into this treatment leads to enhanced tumor regression and cure of established tumors in 60% of treated mice. We show that the mechanisms by which the IDO inhibitor leads to this therapeutic potency include (i) an increment of vaccine-induced tumor-infiltrating effector T cells that is facilitated by anti-OX40 and (ii) a decrease of IDO enzyme activity produced by nontumor cells within the tumor microenvironment that results in enhancement of the specificity and the functionality of vaccine-induced effector T cells. Our findings suggest a translatable strategy to enhance the overall efficacy of cancer immunotherapy. Cancer Immunol Res; 6(2); 201-8. ©2018 AACR.
Collapse
Affiliation(s)
- Zuzana Berrong
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | | | - Shamim Ahmad
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mason Webb
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Eslam Mohamed
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Grigori Okoev
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | | | | | - Rasha Abu Eid
- Georgia Cancer Center, Augusta University, Augusta, Georgia.,The University of Aberdeen Dental School and Hospital, The Institute of Medicine, Medical Sciences and Nutrition, The University of Aberdeen, Scotland, United Kingdom
| | | | - John E Janik
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Samir N Khleif
- Georgia Cancer Center, Augusta University, Augusta, Georgia.
| |
Collapse
|
14
|
Du J, Su S, Li H, Shao J, Meng F, Yang M, Qian H, Zou Z, Qian X, Liu B. Low dose irradiation increases adoptive cytotoxic T lymphocyte migration in gastric cancer. Exp Ther Med 2017; 14:5711-5716. [PMID: 29285113 PMCID: PMC5740708 DOI: 10.3892/etm.2017.5305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/03/2017] [Indexed: 01/09/2023] Open
Abstract
Adoptive cellular immunotherapy (ACI) has been demonstrated to be a promising cancer therapeutic; however, the inefficient migration of adoptive immune cells to tumors is one of the rate-limiting factors of ACI. The present study investigated whether 2 Gy low dose irradiation (LDI) was able to increase the migration of adoptive lymphocytes to gastric cancer cells. Treatment with 2 Gy LDI resulted in marked chemokine (C-X-C motif) ligand 9 (CXCL9) and CXCL10 production from gastric cancer cell lines. A Transwell chamber migration assay demonstrated enhanced transmigration of cytotoxic T lymphocytes to gastric cancer cells following LDI treatment. After 2 Gy LDI application to established gastric carcinoma in nude mice, labeled immune cells were infused by intravenous injection and concentrated fluorescence signals were observed at the tumor sites within the mice, with a peak signal at 8-h LDI. Increased numbers of adoptive T cells at the tumor sites were also observed using flow cytometry. Furthermore, a case study of a patient with metastatic gastric cancer who had received ACI treatment combined with 2 Gy LDI provided further evidence that 2 Gy LDI is able to recruit antitumor effector T cells to tumor sites. Therefore, the ability of 2 Gy LDI to convert tumors into inflamed peripheral tissues may be exploited to overcome obstacles at the effector phase of the antitumor immune response and improve the therapeutic efficacy of immunotherapy.
Collapse
Affiliation(s)
- Juan Du
- The Comprehensive Cancer Center of Drum Tower Hospital, The Affiliated Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Shu Su
- The Comprehensive Cancer Center of Drum Tower Hospital, The Affiliated Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Hongyan Li
- The Comprehensive Cancer Center of Drum Tower Hospital, The Affiliated Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Jie Shao
- The Comprehensive Cancer Center of Drum Tower Hospital, The Affiliated Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Fanyan Meng
- The Comprehensive Cancer Center of Drum Tower Hospital, The Affiliated Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Mi Yang
- The Comprehensive Cancer Center of Drum Tower Hospital, The Affiliated Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Hanqing Qian
- The Comprehensive Cancer Center of Drum Tower Hospital, The Affiliated Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Zhengyun Zou
- The Comprehensive Cancer Center of Drum Tower Hospital, The Affiliated Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Xiaoping Qian
- The Comprehensive Cancer Center of Drum Tower Hospital, The Affiliated Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, The Affiliated Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
15
|
Fotaki G, Jin C, Kerzeli IK, Ramachandran M, Martikainen MM, Karlsson-Parra A, Yu D, Essand M. Cancer vaccine based on a combination of an infection-enhanced adenoviral vector and pro-inflammatory allogeneic DCs leads to sustained antigen-specific immune responses in three melanoma models. Oncoimmunology 2017; 7:e1397250. [PMID: 29399398 PMCID: PMC5790347 DOI: 10.1080/2162402x.2017.1397250] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/04/2017] [Accepted: 10/21/2017] [Indexed: 01/07/2023] Open
Abstract
Autologous patient-derived dendritic cells (DCs) modified ex vivo to present tumor-associated antigens (TAAs) are frequently used as cancer vaccines. However, apart from the stringent logistics in producing DCs on a patient basis, accumulating evidence indicate that ex vivo engineered DCs are poor in migration and in fact do not directly present TAA epitopes to naïve T cells in vivo. Instead, it is proposed that bystander host DCs take up material from vaccine-DCs, migrate and subsequently initiate antitumor T-cell responses. We used mouse models to examine the possibility of using pro-inflammatory allogeneic DCs (alloDCs) to activate host DCs and enable them to promote antigen-specific T-cell immunity. We found that alloDCs were able to initiate host DC activation and migration to draining lymph node leading to T-cell activation. The pro-inflammatory milieu created by alloDCs also led to recruitment of NK cells and neutrophils at the site of injection. Vaccination with alloDCs combined with Ad5M(gp100), an infection-enhanced adenovirus encoding the human melanoma-associated antigen gp100 resulted in generation of CD8+ T cells with a T-cell receptor (TCR) specific for the gp10025-33 epitope (gp100-TCR+). Ad5M(gp100)-alloDC vaccination in combination with transfer of gp100-specific pmel-1 T cells resulted in prolonged survival of B16-F10 melanoma-bearing mice and altered the composition of the tumor microenvironment (TME). We hereby propose that alloDCs together with TAA- or neoepitope-encoding Ad5M can become an “off-the-shelf” cancer vaccine, which can reverse the TME-induced immunosuppression and induce host cellular anti-tumor immune responses in patients without the need of a time-consuming preparation step of autologous DCs.
Collapse
Affiliation(s)
- Grammatiki Fotaki
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Chuan Jin
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Iliana Kyriaki Kerzeli
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Mohanraj Ramachandran
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Minttu-Maria Martikainen
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Alex Karlsson-Parra
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Immunicum AB, Gothenburg Sweden
| | - Di Yu
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Magnus Essand
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
16
|
Yuan R, Zhang S, Yu J, Huang Y, Lu D, Cheng R, Huang S, Ao P, Zheng S, Hood L, Zhu X. Beyond cancer genes: colorectal cancer as robust intrinsic states formed by molecular interactions. Open Biol 2017; 7:rsob.170169. [PMID: 29118272 PMCID: PMC5717345 DOI: 10.1098/rsob.170169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/06/2017] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) has complex pathological features that defy the linear-additive reasoning prevailing in current biomedicine studies. In pursuing a mechanistic understanding behind such complexity, we constructed a core molecular–cellular interaction network underlying CRC and investigated its nonlinear dynamical properties. The hypothesis and modelling method has been developed previously and tested in various cancer studies. The network dynamics reveal a landscape of several attractive basins corresponding to both normal intestinal phenotype and robust tumour subtypes, identified by their different molecular signatures. Comparison between the modelling results and gene expression profiles from patients collected at the second affiliated hospital of Zhejiang University is presented as validation. The numerical ‘driving’ experiment suggests that CRC pathogenesis may depend on pathways involved in gastrointestinal track development and molecules associated with mesenchymal lineage differentiation, such as Stat5, BMP, retinoic acid signalling pathways, Runx and Hox transcription families. We show that the multi-faceted response to immune stimulation and therapies, as well as different carcinogenesis and metastasis routes, can be straightforwardly understood and analysed under such a framework.
Collapse
Affiliation(s)
- Ruoshi Yuan
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Suzhan Zhang
- Key Laboratory of Cancer Prevention and Intervention, Chinese Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang Province 310009, People's Republic of China.,Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, People's Republic of China
| | - Jiekai Yu
- Key Laboratory of Cancer Prevention and Intervention, Chinese Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang Province 310009, People's Republic of China.,Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, People's Republic of China
| | - Yanqin Huang
- Key Laboratory of Cancer Prevention and Intervention, Chinese Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang Province 310009, People's Republic of China.,Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, People's Republic of China
| | - Demin Lu
- Key Laboratory of Cancer Prevention and Intervention, Chinese Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang Province 310009, People's Republic of China.,Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, People's Republic of China
| | - Runtan Cheng
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Sui Huang
- Institute for Systems Biology, 401 Terry Ave. N., Seattle, WA 98109-5234, USA
| | - Ping Ao
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China .,Shanghai Center of Quantitative Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - Shu Zheng
- Key Laboratory of Cancer Prevention and Intervention, Chinese Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang Province 310009, People's Republic of China.,Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, People's Republic of China
| | - Leroy Hood
- Institute for Systems Biology, 401 Terry Ave. N., Seattle, WA 98109-5234, USA
| | - Xiaomei Zhu
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China .,Shanghai Center of Quantitative Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| |
Collapse
|
17
|
Morphine and ketamine inhibit immune function of gastric cancer patients by increasing percentage of CD4(+)CD25(+)Foxp3(+) regulatory T cells in vitro. J Surg Res 2016; 203:306-12. [PMID: 27363637 DOI: 10.1016/j.jss.2016.02.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 02/05/2016] [Accepted: 02/24/2016] [Indexed: 01/18/2023]
Abstract
BACKGROUND There is conflicting evidence regarding effects of anesthetic and analgesic drugs on immune function of cancer patients. This study was designed to observe changes of T cell subpopulations in the gastric cancer (GC) patients and to assess effects of morphine and ketamine on the CD4(+) T cells, CD8(+) T cells, and regulatory T cells (Tregs) populations obtained from the GC patients in vitro. METHODS The peripheral blood samples from 20 GC patients and 20 healthy volunteers were obtained. The peripheral blood mononuclear cells were isolated and incubated in a solution containing phorbol-myristate-acetate and ionomycin (2 μL/mL) in the presence or absence of morphine (50 ng/mL) or different-concentration ketamine (25, 50, and 100 μM). The CD4(+) T cells, CD8(+) T cells, and Tregs were determined using the flow cytometric assay. RESULTS The percentages of CD8(+) T cells were significantly decreased, but the ratio of CD4(+)/CD8(+) T cells and Tregs populations was significantly increased in the GC control group compared with the normal control group (P < 0.05). The ratio of CD4(+)/CD8(+) T cells was significantly increased in the groups M and K3 compared with the control group (P < 0.05) but was significantly decreased in the group K1 compared with the group K3. The percentage of Tregs was significantly increased in the groups M, K1, K2, and K3 compared with the control group. With the increased concentrations, ketamine increased the number of Tregs. CONCLUSIONS GC shifts the balance of CD4(+)/CD8(+) T cells toward CD4(+) T cells and increases the Tregs populations by inducing immune responses. Morphine increases the ratio of CD4(+)/CD8(+) T cells and Tregs populations. Ketamine affects the ratio of CD4(+)/CD8(+) T cells and Tregs populations in a dose-dependent model.
Collapse
|
18
|
Urquiza M, Melo-Cardenas J, Aguillon R, Kipps TJ, Castro JE. Intratumoral injection of Ad-ISF35 (Chimeric CD154) breaks tolerance and induces lymphoma tumor regression. Hum Gene Ther 2015; 26:14-25. [PMID: 25382101 DOI: 10.1089/hum.2014.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Ad-ISF35, an adenovirus vector encoding a membrane-bound engineered CD154 chimeric protein (ISF35), induces complete A20 lymphoma tumor regression in mice after intratumoral direct injection (IDI). Ad-ISF35 induced durable local and systemic antitumor responses associated with a rapid tumor infiltration of macrophages and neutrophils as well as increased levels of proinflammatory cytokines in the tumor microenvironment. Ad-ISF35 IDI transduced preferentially fibroblasts and macrophages present in the tumor microenvironment, and ISF35 protein expression was observed in only 0.25% of cells present in the tumor. Moreover, Ad-ISF35 IDI induced upregulation of CD40 in tumor and immune regulatory cells, including those that did not express ISF35, suggesting the presence of a strong bystander effect. These responses resulted in the generation of IFN-γ-secreting cytotoxic lymphocytes and the production of specific cytotoxic antibodies against lymphoma cells. Overall, cellular immune therapy based on ISF35 induced phenotypic changes in the tumor cells and tumor microenvironment that were associated with a break in tumor immune tolerance and a curative antitumor effect in this lymphoma mouse model. Our data highlight the potential activity that modulation of costimulatory signaling has in cancer therapy.
Collapse
Affiliation(s)
- Mauricio Urquiza
- 1 Moores Cancer Center, University of California-San Diego , La Jolla, CA 92093-0820
| | | | | | | | | |
Collapse
|
19
|
Draghiciu O, Boerma A, Hoogeboom BN, Nijman HW, Daemen T. A rationally designed combined treatment with an alphavirus-based cancer vaccine, sunitinib and low-dose tumor irradiation completely blocks tumor development. Oncoimmunology 2015; 4:e1029699. [PMID: 26451295 PMCID: PMC4589062 DOI: 10.1080/2162402x.2015.1029699] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/09/2015] [Accepted: 03/11/2015] [Indexed: 10/29/2022] Open
Abstract
The clinical efficacy of therapeutic cancer vaccines remains limited. For effective immunotherapeutic responses in cancer patients, multimodal approaches capable of both inducing antitumor immune responses and bypassing tumor-mediated immune escape seem essential. Here, we report on a combination therapy comprising sunitinib (40 mg/kg), single low-dose (14 Gy) tumor irradiation and immunization with a therapeutic cancer vaccine based on a Semliki Forest virus vector encoding the oncoproteins E6 and E7 of human papillomavirus (SFVeE6,7). We previously demonstrated that either low-dose irradiation or sunitinib in single combination with SFVeE6,7 immunizations enhanced the intratumoral ratio of antitumor effector cells to myeloid-derived suppressor cells (MDSCs). On the basis of these results we designed a triple treatment combinatorial regimen. The trimodal sunitinib, low-dose irradiation and SFVeE6,7 immunization therapy resulted in stronger intratumoral MDSC depletion than sunitinib alone. Concomitantly, the highest levels of intratumoral E7-specific CD8+ T cells were attained after triple treatment. Approximately 75% of these cells were positive for the early activation marker CD69. The combination of sunitinib, low-dose tumor irradiation and SFVeE6,7 immunization dramatically changed the intratumoral immune compartment. Whereas control tumors contained 0.02 E7-specific CD8+ T cells per MDSC, triple treatment tumors contained more than 200 E7-specific CD8+ T cells per MDSC, a 10,000-fold increased ratio. As a result, the triple treatment strongly enhanced the immunotherapeutic antitumor effect, blocking tumor development altogether and leading to 100% tumor-free survival of tumor-bearing mice. This study demonstrates that this multimodal approach elicits superior antitumor effects and should be considered for clinical applications.
Collapse
Affiliation(s)
- Oana Draghiciu
- Department of Medical Microbiology; Tumor Virology and Cancer Immunotherapy; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| | - Annemarie Boerma
- Department of Medical Microbiology; Tumor Virology and Cancer Immunotherapy; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| | - Baukje Nynke Hoogeboom
- Department of Medical Microbiology; Tumor Virology and Cancer Immunotherapy; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| | - Hans W Nijman
- Department of Gynecology; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| | - Toos Daemen
- Department of Medical Microbiology; Tumor Virology and Cancer Immunotherapy; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| |
Collapse
|
20
|
Effects of dexmedetomidine on patients undergoing radical gastrectomy. J Surg Res 2015; 194:147-53. [DOI: 10.1016/j.jss.2014.10.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 09/01/2014] [Accepted: 10/03/2014] [Indexed: 01/10/2023]
|
21
|
Schaer DA, Hirschhorn-Cymerman D, Wolchok JD. Targeting tumor-necrosis factor receptor pathways for tumor immunotherapy. J Immunother Cancer 2014; 2:7. [PMID: 24855562 PMCID: PMC4030310 DOI: 10.1186/2051-1426-2-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 03/13/2014] [Indexed: 02/08/2023] Open
Abstract
With the success of ipilimumab and promise of programmed death-1 pathway-targeted agents, the field of tumor immunotherapy is expanding rapidly. Newer targets for clinical development include select members of the tumor necrosis factor receptor (TNFR) family. Agonist antibodies to these co-stimulatory molecules target both T and B cells, modulating T-cell activation and enhancing immune responses. In vitro and in vivo preclinical data have provided the basis for continued development of 4-1BB, OX40, glucocorticoid-induced TNFR-related gene, herpes virus entry mediator, and CD27 as potential therapies for patients with cancer. In this review, we summarize the immune response to tumors, consider preclinical and early clinical data on select TNFR family members, discuss potential translational challenges and suggest possible combination therapies with the aim of inducing durable antitumor responses.
Collapse
Affiliation(s)
- David A Schaer
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA.,Current address: Department of Cancer Immunobiology, ImClone Systems, a wholly-owned subsidiary of Eli Lilly & Co, New York, NY 10016, USA
| | - Daniel Hirschhorn-Cymerman
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Jedd D Wolchok
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA.,Weill Cornell Medical College, New York, NY 10065, USA.,Ludwig Collaborative Lab, New York, NY 10065, USA.,Ludwig Center for Cancer Immunotherapy at Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
22
|
Oikonomopoulou K, Brinc D, Hadjisavvas A, Christofi G, Kyriacou K, Diamandis EP. The bifacial role of helminths in cancer: Involvement of immune and non-immune mechanisms. Crit Rev Clin Lab Sci 2014; 51:138-48. [DOI: 10.3109/10408363.2014.886180] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
23
|
Tao L, Huang G, Shi S, Chen L. Bevacizumab improves the antitumor efficacy of adoptive cytokine-induced killer cells therapy in non-small cell lung cancer models. Med Oncol 2013; 31:777. [DOI: 10.1007/s12032-013-0777-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 11/16/2013] [Indexed: 01/08/2023]
|
24
|
Draghiciu O, Walczak M, Hoogeboom BN, Franken KL, Melief KJ, Nijman HW, Daemen T. Therapeutic immunization and local low-dose tumor irradiation, a reinforcing combination. Int J Cancer 2013; 134:859-72. [DOI: 10.1002/ijc.28418] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 07/03/2013] [Indexed: 11/11/2022]
Affiliation(s)
- Oana Draghiciu
- Department of Medical Microbiology; Molecular Virology Section; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Mateusz Walczak
- Department of Medical Microbiology; Molecular Virology Section; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Baukje Nynke Hoogeboom
- Department of Medical Microbiology; Molecular Virology Section; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Kees L.M.C. Franken
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden The Netherlands
| | - Kees J.M. Melief
- Department of Gynecology; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Hans W. Nijman
- Department of Gynecology; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| | - Toos Daemen
- Department of Medical Microbiology; Molecular Virology Section; University of Groningen, University Medical Center Groningen; Groningen The Netherlands
| |
Collapse
|
25
|
Shi S, Wang R, Chen Y, Song H, Chen L, Huang G. Combining antiangiogenic therapy with adoptive cell immunotherapy exerts better antitumor effects in non-small cell lung cancer models. PLoS One 2013; 8:e65757. [PMID: 23799045 PMCID: PMC3683034 DOI: 10.1371/journal.pone.0065757] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 04/29/2013] [Indexed: 12/14/2022] Open
Abstract
Introduction Cytokine-induced killer cells (CIK cells) are a heterogeneous subset of ex-vivo expanded T lymphocytes which are characterized with a MHC-unrestricted tumor-killing activity and a mixed T-NK phenotype. Adoptive CIK cells transfer, one of the adoptive immunotherapy represents a promising nontoxic anticancer therapy. However, in clinical studies, the therapeutic activity of adoptive CIK cells transfer is not as efficient as anticipated. Possible explanations are that abnormal tumor vasculature and hypoxic tumor microenvironment could impede the infiltration and efficacy of lymphocytes. We hypothesized that antiangiogenesis therapy could improve the antitumor activity of CIK cells by normalizing tumor vasculature and modulating hypoxic tumor microenvironment. Methods We combined recombinant human endostatin (rh-endostatin) and CIK cells in the treatment of lung carcinoma murine models. Intravital microscopy, dynamic contrast enhanced magnetic resonance imaging, immunohistochemistry, and flow cytometry were used to investigate the tumor vasculature and hypoxic microenvironment as well as the infiltration of immune cells. Results Our results indicated that rh-endostatin synergized with adoptive CIK cells transfer to inhibit the growth of lung carcinoma. We found that rh-endostatin normalized tumor vasculature and reduced hypoxic area in the tumor microenvironment. Hypoxia significantly inhibited the proliferation, cytotoxicity and migration of CIK cells in vitro and impeded the homing of CIK cells into tumor parenchyma ex vivo. Furthermore, we found that treatment with rh-endostatin significantly increased the homing of CIK cells and decreased the accumulation of suppressive immune cells in the tumor tissue. In addition, combination therapy produced higher level of tumor-infiltration lymphocytes compared with other treatments. Conclusions Our results demonstrate that rh-endostatin improves the therapeutic effect of adoptive CIK cells therapy against lung carcinomas and unmask the mechanisms of the synergistic antitumor efficacy, providing a new rationale for combining antiangiogenesis therapy with immunotherapy in the treatment of lung cancer.
Collapse
Affiliation(s)
- Shujing Shi
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
| | - Rui Wang
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
| | - Yitian Chen
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
| | - Haizhu Song
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
| | - Longbang Chen
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
- * E-mail: (GCH); (LBC)
| | - Guichun Huang
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
- * E-mail: (GCH); (LBC)
| |
Collapse
|
26
|
Shapira-Frommer R, Schachter J. Adoptive immunotherapy of advanced melanoma. Curr Treat Options Oncol 2012; 13:340-53. [PMID: 22864561 DOI: 10.1007/s11864-012-0203-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adoptive cell therapy (ACT) has emerged as an effective therapy for patients with metastatic melanoma. Since the first introduction of the protocol in 1988 [1], major improvements have been achieved with response rates of 40%-72% among patients who were resistant to previous treatment lines. Both cell product and conditioning regimen are major determinants of treatment efficacy; therefore, developing ACT protocols explore diverse ways to establish autologous intra-tumoral lymphocyte cultures or peripheral effector cells as well as different lymphodepleting regimens. While a proof of feasibility and a proof of concept had been established with previous published results, ACT will need to move beyond single-center experiences, to confirmatory, multi-center studies. If ACT is to move into widespread practice, it will be necessary to develop reproducible high quality cell production methods and accepted lymphodepleting regimen. Two new drugs, ipilimumab (Yervoy, Bristol-Myers Squibb) and vemurafenib (Zelboraf, Roche), were approved in 2011 for the treatment of metastatic melanoma based on positive phase III trials. Both drugs show a clear overall survival benefit, so the timing of when to use ACT will need to be carefully thought out. In contrast to these 2 new, commercially available outpatient treatments, ACT is a personally-specified product and labor-intensive therapy that demands both acquisition of high standard laboratory procedures and close clinical inpatient monitoring during treatment. It is unique among other anti-melanoma treatments, providing the potential for a durable response following a single, self-limited treatment. This perspective drives the efforts to make this protocol accessible for more patients and to explore modifications that may optimize treatment results.
Collapse
Affiliation(s)
- Ronnie Shapira-Frommer
- Ella Institute for the Treatment and Research of Melanoma, Sheba Medical Center, Ramat-Gan, 52621, Israel.
| | | |
Collapse
|
27
|
Myelopotentiating effect of curcumin in tumor-bearing host: Role of bone marrow resident macrophages. Toxicol Appl Pharmacol 2012; 263:111-21. [DOI: 10.1016/j.taap.2012.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/28/2012] [Accepted: 06/05/2012] [Indexed: 12/16/2022]
|