1
|
Chávez-Pacheco JL, Castillejos-López M, Hernández-Regino LM, Velasco-Hidalgo L, Zapata-Tarres M, Correa-Carranza V, Rosario-Méndez G, Barrientos-Ríos R, Aquino-Gálvez A, Torres-Espíndola LM. Challenges in Treating Pediatric Cancer Patients during the COVID-19 Pandemic: Balancing Risks and Care. Viruses 2024; 16:690. [PMID: 38793571 PMCID: PMC11125850 DOI: 10.3390/v16050690] [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: 03/14/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
The COVID-19 pandemic has resulted in millions of fatalities worldwide. The case of pediatric cancer patients stands out since, despite being considered a population at risk, few studies have been carried out concerning symptom detection or the description of the mechanisms capable of modifying the course of the COVID-19 disease, such as the interaction and response between the virus and the treatment given to cancer patients. By synthesizing existing studies, this paper aims to expose the treatment challenges for pediatric patients with COVID-19 in an oncology context. Additionally, this updated review includes studies that utilized the antiviral agents Remdesivir and PaxlovidTM in pediatric cancer patients. There is no specific treatment designed exclusively for pediatric cancer patients dealing with COVID-19, and it is advisable to avoid self-medication to prevent potential side effects. Managing COVID-19 in pediatric cancer patients is indeed a substantial challenge. New strategies, such as chemotherapy application rooms, have been implemented for children with cancer who were positive for COVID-19 but asymptomatic since the risk of disease progression is greater than the risk of complications from SARS-CoV-2.
Collapse
Affiliation(s)
- Juan Luis Chávez-Pacheco
- Pharmacology Laboratory, National Institute of Pediatrics, Mexico City 04530, Mexico; (J.L.C.-P.); (L.M.H.-R.); (V.C.-C.); (G.R.-M.)
| | - Manuel Castillejos-López
- Epidemiology and Infectology, National Institute of Respiratory Diseases, Mexico City 14080, Mexico;
| | - Laura M. Hernández-Regino
- Pharmacology Laboratory, National Institute of Pediatrics, Mexico City 04530, Mexico; (J.L.C.-P.); (L.M.H.-R.); (V.C.-C.); (G.R.-M.)
| | | | - Marta Zapata-Tarres
- Head of Research Coordination at Mexican Social Security Institute Foundation, Mexico City 06600, Mexico;
| | - Valeria Correa-Carranza
- Pharmacology Laboratory, National Institute of Pediatrics, Mexico City 04530, Mexico; (J.L.C.-P.); (L.M.H.-R.); (V.C.-C.); (G.R.-M.)
| | - Guillermo Rosario-Méndez
- Pharmacology Laboratory, National Institute of Pediatrics, Mexico City 04530, Mexico; (J.L.C.-P.); (L.M.H.-R.); (V.C.-C.); (G.R.-M.)
| | - Rehotbevely Barrientos-Ríos
- Cytogenetics Laboratory, Department of Human Genetics, National Institute of Pediatrics, Mexico City 04530, Mexico;
| | - Arnoldo Aquino-Gálvez
- Molecular Biology Laboratory, Pulmonary Fibrosis Department, National Institute of Respiratory Diseases, Mexico City 14080, Mexico
| | - Luz María Torres-Espíndola
- Pharmacology Laboratory, National Institute of Pediatrics, Mexico City 04530, Mexico; (J.L.C.-P.); (L.M.H.-R.); (V.C.-C.); (G.R.-M.)
| |
Collapse
|
2
|
Stepanenko AA, Sosnovtseva AO, Valikhov MP, Chernysheva AA, Abramova OV, Naumenko VA, Chekhonin VP. The need for paradigm shift: prognostic significance and implications of standard therapy-related systemic immunosuppression in glioblastoma for immunotherapy and oncolytic virotherapy. Front Immunol 2024; 15:1326757. [PMID: 38390330 PMCID: PMC10881776 DOI: 10.3389/fimmu.2024.1326757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Despite significant advances in our knowledge regarding the genetics and molecular biology of gliomas over the past two decades and hundreds of clinical trials, no effective therapeutic approach has been identified for adult patients with newly diagnosed glioblastoma, and overall survival remains dismal. Great hopes are now placed on combination immunotherapy. In clinical trials, immunotherapeutics are generally tested after standard therapy (radiation, temozolomide, and steroid dexamethasone) or concurrently with temozolomide and/or steroids. Only a minor subset of patients with progressive/recurrent glioblastoma have benefited from immunotherapies. In this review, we comprehensively discuss standard therapy-related systemic immunosuppression and lymphopenia, their prognostic significance, and the implications for immunotherapy/oncolytic virotherapy. The effectiveness of immunotherapy and oncolytic virotherapy (viro-immunotherapy) critically depends on the activity of the host immune cells. The absolute counts, ratios, and functional states of different circulating and tumor-infiltrating immune cell subsets determine the net immune fitness of patients with cancer and may have various effects on tumor progression, therapeutic response, and survival outcomes. Although different immunosuppressive mechanisms operate in patients with glioblastoma/gliomas at presentation, the immunological competence of patients may be significantly compromised by standard therapy, exacerbating tumor-related systemic immunosuppression. Standard therapy affects diverse immune cell subsets, including dendritic, CD4+, CD8+, natural killer (NK), NKT, macrophage, neutrophil, and myeloid-derived suppressor cell (MDSC). Systemic immunosuppression and lymphopenia limit the immune system's ability to target glioblastoma. Changes in the standard therapy are required to increase the success of immunotherapies. Steroid use, high neutrophil-to-lymphocyte ratio (NLR), and low post-treatment total lymphocyte count (TLC) are significant prognostic factors for shorter survival in patients with glioblastoma in retrospective studies; however, these clinically relevant variables are rarely reported and correlated with response and survival in immunotherapy studies (e.g., immune checkpoint inhibitors, vaccines, and oncolytic viruses). Our analysis should help in the development of a more rational clinical trial design and decision-making regarding the treatment to potentially improve the efficacy of immunotherapy or oncolytic virotherapy.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasiia O. Sosnovtseva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marat P. Valikhov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasia A. Chernysheva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga V. Abramova
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Victor A. Naumenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
3
|
Gao Z, Azar J, Zhu H, Williams-Perez S, Kang SW, Marginean C, Rubinstein MP, Makawita S, Lee HS, Camp ER. Translational and oncologic significance of tertiary lymphoid structures in pancreatic adenocarcinoma. Front Immunol 2024; 15:1324093. [PMID: 38361928 PMCID: PMC10867206 DOI: 10.3389/fimmu.2024.1324093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/11/2024] [Indexed: 02/17/2024] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is an aggressive tumor with poor survival and limited treatment options. PDAC resistance to immunotherapeutic strategies is multifactorial, but partially owed to an immunosuppressive tumor immune microenvironment (TiME). However, the PDAC TiME is heterogeneous and harbors favorable tumor-infiltrating lymphocyte (TIL) populations. Tertiary lymphoid structures (TLS) are organized aggregates of immune cells that develop within non-lymphoid tissue under chronic inflammation in multiple contexts, including cancers. Our current understanding of their role within the PDAC TiME remains limited; TLS are complex structures with multiple anatomic features such as location, density, and maturity that may impact clinical outcomes such as survival and therapy response in PDAC. Similarly, our understanding of methods to manipulate TLS is an actively developing field of research. TLS may function as anti-tumoral immune niches that can be leveraged as a therapeutic strategy to potentiate both existing chemotherapeutic regimens and potentiate future immune-based therapeutic strategies to improve patient outcomes. This review seeks to cover anatomy, relevant features, immune effects, translational significance, and future directions of understanding TLS within the context of PDAC.
Collapse
Affiliation(s)
- Zachary Gao
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Joseph Azar
- The Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Huili Zhu
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Sophia Williams-Perez
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Sung Wook Kang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Celia Marginean
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Mark P. Rubinstein
- The Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Shalini Makawita
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Hyun-Sung Lee
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - E. Ramsay Camp
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX, United States
| |
Collapse
|
4
|
Yu H, Chen J, Zhou Y, Sheng J, Zhang X, He L, Chen L, Chu Q, Zhang L, Hong S. Impact of prophylactic dexamethasone on the efficacy of immune checkpoint inhibitors plus platinum-based chemotherapy in patients with advanced Non-Squamous Non-Small-Cell lung cancer. Int Immunopharmacol 2023; 125:111138. [PMID: 37948858 DOI: 10.1016/j.intimp.2023.111138] [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: 05/14/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Baseline corticosteroids exposure is associated with inferior clinical outcomes in patients with non-small-cell lung cancer (NSCLC) treated with programmed cell death-1 (PD-1) axis blockade. Dexamethasone is a potent corticosteroid used in the prevention of chemotherapy-associated adverse events (CAAEs). OBJECTIVE Since dexamethasone has immunosuppressive properties, this study attempted to elucidate its effects on the efficacy of immunotherapy plus chemotherapy in patients with non-squamous NSCLC. METHODS The study retrospectively analyzed the medical records of 254 advanced non-squamous NSCLC patients who received front-line treatment with a PD-1 pathway inhibitor and platinum-based chemotherapy at three academic institutions. The average dosage of prophylactic dexamethasone per chemotherapy cycle was calculated. Patients were divided into three groups based on the dose of dexamethasone: High-d (≥24 mg), Moderate-d (12-24 mg), and Low-d (<12 mg). Spearman's rank correlation was used to assess the correlation between the dosage of dexamethasone and progression-free survival (PFS). Logistic regression was used to assess the correlation between dexamethasone dosage and the occurrence of immune related adverse effects (irAE). Univariate and multivariate Cox proportional hazards regression models were used to analyze the differences in survival among the different dexamethasone dosage groups. RESULT The dosage of prophylactic dexamethasone was not significantly correlated with PFS (Spearman's rho = -0.103, P = 0.098). Results from the univariate [hazard ratio (HR)Low-d/High-d, 1.00; P = 0.997; HRModerate-d/High-d, 0.85; P = 0.438] and multivariate (HRLow-d/High-d, 0.71; P = 0.174; HRModerate-d/High-d, 0.87; P = 0.512) analyses showed no significant association between dexamethasone and PFS. Dexamethasone did not have significant effect on the objective response rate, disease control rate or overall survival. The toxicity profiles of irAE were similar across all three groups. CONCLUSION The results of this study suggest that the use of prophylactic dexamethasone does not have an adverse effect on the clinical outcomes of non-squamous NSCLC patients treated with PD-1 blockade therapy and chemotherapy. Routine use of dexamethasone for preventing CAAEs should be recommended for patients undergoing combined immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Hui Yu
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Jingyu Chen
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yixin Zhou
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Jin Sheng
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Zhejiang 310018, Hangzhou, China
| | - Xuanye Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Lina He
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Likun Chen
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China.
| | - Shaodong Hong
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China.
| |
Collapse
|
5
|
Zou X, Lin X, Cheng H, Chen Y, Wang R, Ma M, Liu Y, Dai Z, Tasiheng Y, Yan Y, Hou Q, Ding F, Chen H, Yu X, Wang X, Liu C. Characterization of intratumoral tertiary lymphoid structures in pancreatic ductal adenocarcinoma: cellular properties and prognostic significance. J Immunother Cancer 2023; 11:e006698. [PMID: 37364934 PMCID: PMC10410893 DOI: 10.1136/jitc-2023-006698] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Tumor-associated tertiary lymphoid structures (TLSs) are functional immune-responsive niches that are not fully understood in pancreatic ductal adenocarcinoma (PDAC). METHODS Fluorescent multiplex immunohistochemistry was performed on sequential sections of surgically resected tumor tissues from 380 PDAC patients without preoperative treatment (surgery alone (SA)) and 136 patients pretreated with neoadjuvant treatment (NAT). Multispectral images were processed via machine learning and image processing platforms, inForm V.2.4 and HALO V.3.2; TLS regions were segmented, and the cells were identified and quantified. The cellular composition and immunological properties of TLSs and their adjacent tissues in PDAC were scored and compared, and their association with prognosis was further examined. RESULTS Intratumoral TLSs were identified in 21.1% (80/380) of patients in the SA group and 15.4% (21/136) of patients in the NAT group. In the SA group, the presence of intratumoral TLSs was significantly associated with improved overall survival (OS) and progression-free survival. The existence of intratumoral TLSs was correlated with elevated levels of infiltrating CD8+T, CD4+T, B cells and activated immune cells in adjacent tissues. A nomogram model was generated with TLS presence as a variable, which successfully predicted PDAC patient OS in an external validation cohort (n=123). In the NAT group, samples exhibited a lower proportion of B cells and a higher proportion of regulatory T cells within intratumoral TLSs. Additionally, these TLSs were smaller in size, with a lower overall maturation level and reduced immune cell activation, and the prognostic value of TLS presence was insignificant in the NAT cohort. CONCLUSION Our study systematically revealed the cellular properties and prognostic values of intratumoral TLSs in PDAC and described the potential impact of NAT on TLS development and function.
Collapse
Affiliation(s)
- Xuan Zou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Xuan Lin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - He Cheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yusheng Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Ruijie Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Mingjian Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yu Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Zhengjie Dai
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yesboli Tasiheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yu Yan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Qinqin Hou
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Fei Ding
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Huan Chen
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Xu Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Radiation Oncology, Cancer Research Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, People's Republic of China
| | - Chen Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
6
|
Murphy L, Shaker J, Buggy DJ. Anaesthetic Techniques and Strategies: Do They Influence Oncological Outcomes? Curr Oncol 2023; 30:5309-5321. [PMID: 37366886 PMCID: PMC10296968 DOI: 10.3390/curroncol30060403] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND With the global disease burden of cancer increasing, and with at least 60% of cancer patients requiring surgery and, hence, anaesthesia over their disease course, the question of whether anaesthetic and analgesia techniques during primary cancer resection surgery might influence long term oncological outcomes assumes high priority. METHODS We searched the available literature linking anaesthetic-analgesic techniques and strategies during tumour resection surgery to oncological outcomes and synthesised this narrative review, predominantly using studies published since 2019. Current evidence is presented around opioids, regional anaesthesia, propofol total intravenous anaesthesia (TIVA) and volatile anaesthesia, dexamethasone, dexmedetomidine, non-steroidal anti-inflammatory medications and beta-blockers. CONCLUSIONS The research base in onco-anaesthesia is expanding. There continue to be few sufficiently powered RCTs, which are necessary to confirm a causal link between any perioperative intervention and long-term oncologic outcome. In the absence of any convincing Level 1 recommending a change in practice, long-term oncologic benefit should not be part of the decision on choice of anaesthetic technique for tumour resection surgery.
Collapse
Affiliation(s)
- Liam Murphy
- Department of Anaesthesiology & Perioperative Medicine, Mater University Hospital, School of Medicine, University College Dublin, D07 R2WY Dublin, Ireland; (J.S.); (D.J.B.)
| | - John Shaker
- Department of Anaesthesiology & Perioperative Medicine, Mater University Hospital, School of Medicine, University College Dublin, D07 R2WY Dublin, Ireland; (J.S.); (D.J.B.)
| | - Donal J. Buggy
- Department of Anaesthesiology & Perioperative Medicine, Mater University Hospital, School of Medicine, University College Dublin, D07 R2WY Dublin, Ireland; (J.S.); (D.J.B.)
- European Society of Anaesthesiology and Intensive Care Onco-Anaesthesiology Research Group, 24 Rue des Comédiens, B-1000 Brussels, Belgium
- Outcomes Research, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
7
|
Effect of Dexamethasone on Abiraterone Pharmacokinetics in Mice: Determined by LC/MS Analysis. MEDICINES 2023; 10:medicines10030021. [PMID: 36976310 PMCID: PMC10053955 DOI: 10.3390/medicines10030021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Background: Abiraterone acetate is a cytochrome P450 17A1 (CYP17A1) inhibitor that is indicated for use in both castration-resistant and castration-sensitive prostate cancer patients. To manage the mineralocorticoid effects of CYP17A1 inhibition, a glucocorticoid such as dexamethasone is co-administered with abiraterone. The goal of the present study was to understand the effect of dexamethasone on the disposition of abiraterone. Methods: Adult male CD-1 mice were treated with either dexamethasone (80 mg/kg/day) or vehicle for three consecutive days, followed by the administration of a single dose of abiraterone acetate (180 mg/kg) as an oral gavage. Blood samples were collected by tail bleeding at timepoints between 0 to 24 h. Subsequently, abiraterone was extracted from the mouse serum using a neutral pH condition and serum abiraterone levels were determined using a liquid chromatography–mass spectrometry assay. Results: Our results demonstrated that dexamethasone lowered the maximum plasma concentration and area under the curve parameters by approximately five- and ten-fold, respectively. Similar effects were also observed on the plasma half-life and oral clearance parameters. This is the first report of dexamethasone effect on abiraterone disposition in vivo. Conclusions: We conclude that dexamethasone has the potential to reduce the plasma abiraterone level and thus compromise its CYP17A1 inhibitory ability in the procancerous androgen biosynthesis pathway. Thus, use of a higher abiraterone dose may be warranted when used alongside dexamethasone.
Collapse
|
8
|
Karra AG, Tsialtas I, Kalousi FD, Georgantopoulos A, Sereti E, Dimas K, Psarra AMG. Increased Expression of the Mitochondrial Glucocorticoid Receptor Enhances Tumor Aggressiveness in a Mouse Xenograft Model. Int J Mol Sci 2023; 24:ijms24043740. [PMID: 36835152 PMCID: PMC9966287 DOI: 10.3390/ijms24043740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Mitochondria are important organelles for cellular physiology as they generate most of the energy requirements of the cell and orchestrate many biological functions. Dysregulation of mitochondrial function is associated with many pathological conditions, including cancer development. Mitochondrial glucocorticoid receptor (mtGR) is proposed as a crucial regulator of mitochondrial functions via its direct involvement in the regulation of mitochondrial transcription, oxidative phosphorylation (OXPHOS), enzymes biosynthesis, energy production, mitochondrial-dependent apoptosis, and regulation of oxidative stress. Moreover, recent observations revealed the interaction of mtGR with the pyruvate dehydrogenase (PDH), a key player in the metabolic switch observed in cancer, indicating direct involvement of mtGR in cancer development. In this study, by using a xenograft mouse model of mtGR-overexpressing hepatocarcinoma cells, we showed increased mtGR-associated tumor growth, which is accompanied by reduced OXPHOS biosynthesis, reduction in PDH activity, and alterations in the Krebs cycle and glucose metabolism, metabolic alterations similar to those observed in the Warburg effect. Moreover, autophagy activation is observed in mtGR-associated tumors, which further support tumor progression via increased precursors availability. Thus, we propose that increased mitochondrial localization of mtGR is associated with tumor progression possible via mtGR/PDH interaction, which could lead to suppression of PDH activity and modulation of mtGR-induced mitochondrial transcription that ends up in reduced OXPHOS biosynthesis and reduced oxidative phosphorylation versus glycolytic pathway energy production, in favor of cancer cells.
Collapse
Affiliation(s)
- Aikaterini G. Karra
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Ioannis Tsialtas
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Foteini D. Kalousi
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Achilleas Georgantopoulos
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Evangelia Sereti
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Konstantinos Dimas
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Anna-Maria G. Psarra
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
- Correspondence: ; Tel.: +30-24-1056-5221; Fax: +30-24-1056-5290
| |
Collapse
|
9
|
Dinarello A, Mills TS, Tengesdal IW, Powers NE, Azam T, Dinarello CA. Dexamethasone and OLT1177 Cooperate in the Reduction of Melanoma Growth by Inhibiting STAT3 Functions. Cells 2023; 12:294. [PMID: 36672229 PMCID: PMC9856388 DOI: 10.3390/cells12020294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
The NLRP3 inflammasome is a multimolecular complex that processes inactive IL-1β and IL-18 into proinflammatory cytokines. OLT1177 is an orally active small compound that specifically inhibits NLRP3. Here, B16F10 melanoma were implanted in mice and treated with OLT1177 as well as combined with the glucocorticoid dexamethasone. At sacrifice, OLT1177 treated mice had significantly smaller tumors compared to tumor-bearing mice treated with vehicle. However, the combined treatment of OLT1177 plus dexamethasone revealed a greater suppression of tumor growth. This reduction was accompanied by a downregulation of nuclear and mitochondrial STAT3-dependent gene transcription and by a significant reduction of STAT3 Y705 and S727 phosphorylations in the tumors. In vitro, the human melanoma cell line 1205Lu, stimulated with IL-1α, exhibited significantly lower levels of STAT3 Y705 phosphorylation by the combination treatment, thus affecting the nuclear functions of STAT3. In the same cells, STAT3 serine 727 phosphorylation was also lower, affecting the mitochondrial functions of STAT3. In addition, metabolic analyses revealed a marked reduction of ATP production rate and glycolytic reserve in cells treated with the combination of OLT1177 plus dexamethasone. These findings demonstrate that the combination of OLT1177 and dexamethasone reduces tumor growth by targeting nuclear as well as mitochondrial functions of STAT3.
Collapse
Affiliation(s)
- Alberto Dinarello
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
| | - Taylor S. Mills
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
| | - Isak W. Tengesdal
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Nicholas E. Powers
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
| | - Tania Azam
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
10
|
Willems M, Scherpereel A, Wasielewski E, Raskin J, Brossel H, Fontaine A, Grégoire M, Halkin L, Jamakhani M, Heinen V, Louis R, Duysinx B, Hamaidia M, Willems L. Excess of blood eosinophils prior to therapy correlates with worse prognosis in mesothelioma. Front Immunol 2023; 14:1148798. [PMID: 37026006 PMCID: PMC10070849 DOI: 10.3389/fimmu.2023.1148798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 02/20/2023] [Indexed: 04/08/2023] Open
Abstract
Background Only a fraction of patients with malignant pleural mesothelioma (MPM) will respond to chemo- or immunotherapy. For the majority, the condition will irremediably relapse after 13 to 18 months. In this study, we hypothesized that patients' outcome could be correlated to their immune cell profile. Focus was given to peripheral blood eosinophils that, paradoxically, can both promote or inhibit tumor growth depending on the cancer type. Methods The characteristics of 242 patients with histologically proven MPM were retrospectively collected in three centers. Characteristics included overall survival (OS), progression-free survival (PFS), overall response rate (ORR) and disease control rate (DCR). The mean absolute eosinophil counts (AEC) were determined by averaging AEC data sets of the last month preceding the administration of chemo- or immunotherapy. Results An optimal cutoff of 220 eosinophils/µL of blood segregated the cohort into two groups with significantly different median OS after chemotherapy (14 and 29 months above and below the threshold, p = 0.0001). The corresponding two-year OS rates were 28% and 55% in the AEC ≥ 220/µL and AEC < 220/µL groups, respectively. Based on shorter median PFS (8 vs 17 months, p < 0.0001) and reduced DCR (55.9% vs 35.2% at 6 months), the response to standard chemotherapy was significantly affected in the AEC ≥ 220/µL subset. Similar conclusions were also drawn from data sets of patients receiving immune checkpoint-based immunotherapy. Conclusion In conclusion, baseline AEC ≥ 220/µL preceding therapy is associated with worse outcome and quicker relapse in MPM.
Collapse
Affiliation(s)
- Mégane Willems
- Laboratory of Molecular and Cellular Epigenetics (GIGA at University of Liege), Sart-Tilman, Molecular Biology, Teaching and Research Centre (TERRA), Gembloux, Belgium
| | - Arnaud Scherpereel
- Department of Pneumology and Thoracic Oncology, (CHU Lille) and INSERM (ONCOTHAI), Lille, France
| | - Eric Wasielewski
- Department of Pneumology and Thoracic Oncology, (CHU Lille) and INSERM (ONCOTHAI), Lille, France
| | - Jo Raskin
- Department of Pulmonology and Thoracic Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Hélène Brossel
- Laboratory of Molecular and Cellular Epigenetics (GIGA at University of Liege), Sart-Tilman, Molecular Biology, Teaching and Research Centre (TERRA), Gembloux, Belgium
| | - Alexis Fontaine
- Laboratory of Molecular and Cellular Epigenetics (GIGA at University of Liege), Sart-Tilman, Molecular Biology, Teaching and Research Centre (TERRA), Gembloux, Belgium
| | - Mélanie Grégoire
- Laboratory of Molecular and Cellular Epigenetics (GIGA at University of Liege), Sart-Tilman, Molecular Biology, Teaching and Research Centre (TERRA), Gembloux, Belgium
| | - Louise Halkin
- Laboratory of Molecular and Cellular Epigenetics (GIGA at University of Liege), Sart-Tilman, Molecular Biology, Teaching and Research Centre (TERRA), Gembloux, Belgium
| | - Majeed Jamakhani
- Laboratory of Molecular and Cellular Epigenetics (GIGA at University of Liege), Sart-Tilman, Molecular Biology, Teaching and Research Centre (TERRA), Gembloux, Belgium
| | - Vincent Heinen
- Department of Pneumology, University Hospital of Liege, Liege, Belgium
| | - Renaud Louis
- Department of Pneumology, University Hospital of Liege, Liege, Belgium
| | - Bernard Duysinx
- Department of Pneumology, University Hospital of Liege, Liege, Belgium
| | - Malik Hamaidia
- Laboratory of Molecular and Cellular Epigenetics (GIGA at University of Liege), Sart-Tilman, Molecular Biology, Teaching and Research Centre (TERRA), Gembloux, Belgium
| | - Luc Willems
- Laboratory of Molecular and Cellular Epigenetics (GIGA at University of Liege), Sart-Tilman, Molecular Biology, Teaching and Research Centre (TERRA), Gembloux, Belgium
- *Correspondence: Luc Willems,
| |
Collapse
|
11
|
Khadka S, Druffner SR, Duncan BC, Busada JT. Glucocorticoid regulation of cancer development and progression. Front Endocrinol (Lausanne) 2023; 14:1161768. [PMID: 37143725 PMCID: PMC10151568 DOI: 10.3389/fendo.2023.1161768] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/28/2023] [Indexed: 05/06/2023] Open
Abstract
Glucocorticoids are steroid hormones that regulate a host of cellular and physiological functions. However, they are arguably best known for their potent anti-inflammatory properties. Chronic inflammation is well-known to promote the development and progression of numerous types of cancer, and emerging evidence suggests that glucocorticoid regulation of inflammation affects cancer development. However, the timing, intensity, and duration of glucocorticoid signaling have important but often contradictory effects on cancer development. Moreover, glucocorticoids are widely used in parallel with radiation and chemotherapy to control pain, dyspnea, and swelling, but their use may compromise anti-tumor immunity. This review will explore the effects of glucocorticoids on cancer development and progression with particular focus on pro and anti-tumor immunity.
Collapse
|
12
|
As time goes by–developments in surgery for esophageal cancer in the new millennium. Eur Surg 2022. [DOI: 10.1007/s10353-022-00752-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Summary
Background
In the last two decades, both treatment options and epidemiological features of cancer have changed. We studied the influence of related parameters on the outcome of patients undergoing resection for esophageal carcinoma.
Methods
We analyzed 499 consecutive patients who underwent esophagectomy for carcinoma since January 2000, comparing 2000–2010 with 2011–2021 and examining changes over time.
Results
The percentage of men (87.9 vs. 86.9%; p = 0.74) in the two groups was unchanged, whereas mean age increased significantly from 60.8 to 65.2 years (p = 0.000). There was a trend towards an increase of adenocarcinoma (gamma = 0.120, ASE = 0.055). Despite significantly increasing use of induction chemoradiotherapy (p = 0.000) from 7.14% in 2000 to 68.9% in 2021 the distribution of pT, pN stage, grading and the rate of positive lateral resection margins remained unchanged. When comparing the two periods, the overall 30-day mortality was 4.4 vs. 4.2% (p = 0.56), recurrence-free survival was 36.9 vs. 38% at 60 months and 33.9 vs. 36.4% at 120 months (p = 0.93). Tumor-associated survival was 41.1 vs. 45% at 60 months and 35.5 vs. 38.7% at 120 months (p = 0.78). None of the survival rates differed significantly. A multivariable analysis of year of surgery, age, sex, histological subtype, grading, pT, pN, lateral resection margin, and induction therapy showed that only higher pT (p = 0.01), positive pN (p = 0.000), positive lateral margin (p = 0.003), squamous cell carcinoma (p = 0.04) and higher grading (p = 0.026) had a statistically significant, independent, negative influence on prognosis.
Conclusion
Optimized noninvasive and invasive therapeutic modalities have produced only marginal improvement in the prognosis of esophageal cancer within the last two decades.
Collapse
|
13
|
Das T, Mukhopadhyay C. Computational studies suggest compounds restoring function of p53 cancer mutants can bind SARS-CoV-2 spike protein. J Biomol Struct Dyn 2022; 41:3368-3381. [PMID: 35333136 DOI: 10.1080/07391102.2022.2048081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
It is reasonable to think that cancer patients undergoing chemotherapy or immunotherapy may have a more aggressive course if they are positive for the novel coronavirus disease. Their compulsive condition requires investigation into effective drugs. We applied computational techniques to a series of compounds known for restoring the function of p53 cancer mutant p53R175H and p53G245S. Two potent inhibitors, 1-(3-chlorophenyl)-3-(1, 3 -thiazol-2-yl) urea (CTU, PubChem NSC321792) with the highest binding affinity -6.92 kcal/mol followed by a thiosemicarbazone compound N'-(1-(Pyridin-2-yl)ethylidene) azetidine - 1 -carbothiohydrazide (NPC, PubChem NSC319726) with -6.75 kcal/mol were subjected to Molecular Dynamics simulation with receptor binding domain (RBD) and compared with control ligand dexamethasone. In particular, CTU adheres to pocket 1 with an average free energy of binding -21.65 ± 2.89 kcal/mol at the RBD - angiotensin-converting enzyme 2 binding region with the highest frequency of amino acid residues after reaching a local equilibrium in 100 ns MD simulation trajectory. A significant enthalpy contribution from the independent simulations unfolds the possibility of dual binding sites for NPC as shifted pocket 1 (-15.59 ± 5.98 kcal/mol) and pocket 2 (-18.90 ± 5.02 kcal/mol). The obtained results for these two compounds are in good agreement with dexamethasone (-18.45 ± 2.42 kcal/mol). Taken together our findings could facilitate the discovery of small molecules that restore the function of p53 cancer mutants newly against COVID-19 in cancer patients.
Collapse
Affiliation(s)
- Tanushree Das
- Department of Chemistry, University of Calcutta, Kolkata, India
| | | |
Collapse
|
14
|
Tang S, Zhang Z, Oakley RH, Li W, He W, Xu X, Ji M, Xu Q, Chen L, Wellman AS, Li Q, Li L, Li JL, Li X, Cidlowski JA, Li X. Intestinal epithelial glucocorticoid receptor promotes chronic inflammation-associated colorectal cancer. JCI Insight 2021; 6:151815. [PMID: 34784298 PMCID: PMC8783679 DOI: 10.1172/jci.insight.151815] [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: 05/26/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
Synthetic immunosuppressive glucocorticoids (GCs) are widely used to control inflammatory bowel disease (IBD). However, the impact of GC signaling on intestinal tumorigenesis remains controversial. Here, we report that intestinal epithelial GC receptor (GR), but not whole intestinal tissue GR, promoted chronic intestinal inflammation-associated colorectal cancer in both humans and mice. In patients with colorectal cancer, GR was enriched in intestinal epithelial cells and high epithelial cell GR levels were associated with poor prognosis. Consistently, intestinal epithelium–specific deletion of GR (GR iKO) in mice increased macrophage infiltration, improved tissue recovery, and enhanced antitumor response in a chronic inflammation–associated colorectal cancer model. Consequently, GR iKO mice developed fewer and less advanced tumors than control mice. Furthermore, oral GC administration in the early phase of tissue injury delayed recovery and accelerated the formation of aggressive colorectal cancers. Our study reveals that intestinal epithelial GR signaling repressed acute colitis but promoted chronic inflammation–associated colorectal cancer. Our study suggests that colorectal epithelial GR could serve as a predictive marker for colorectal cancer risk and prognosis. Our findings further suggest that, although synthetic GC treatment for IBD should be used with caution, there is a therapeutic window for GC therapy during colorectal cancer development in immunocompetent patients.
Collapse
Affiliation(s)
- Shuang Tang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhan Zhang
- Central for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | | | - Wenling Li
- Biostatistics and Computational Biology Branch, NIEHS/NIH, Research Triangle Park, United States of America
| | - Weijing He
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaojiang Xu
- Integrated Bioinformatics, NIEHS/NIH, Research Triangle Park, United States of America
| | - Ming Ji
- Signal Transduction Laboratory, NIEHS/NIH, Research Triangle Park, United States of America
| | - Qing Xu
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, United States of America
| | - Liang Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Alicia S Wellman
- Signal Transduction Laboratory, NIEHS/NIH, Research Triangle Park, United States of America
| | - Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Leping Li
- Biostatistics and Computational Biology Branch, NIEHS/NIH, Research Triangle Park, United States of America
| | - Jian-Liang Li
- NIEHS/NIH, Research Triangle Park, United States of America
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - John A Cidlowski
- Signal Transduction Laboratory, NIEHS/NIH, Research Triangle Park, United States of America
| | - Xiaoling Li
- Signal Transduction Laboratory, NIEHS/NIH, Research Triangle Park, United States of America
| |
Collapse
|
15
|
Lee Y. Roles of circadian clocks in cancer pathogenesis and treatment. Exp Mol Med 2021; 53:1529-1538. [PMID: 34615982 PMCID: PMC8568965 DOI: 10.1038/s12276-021-00681-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/29/2021] [Accepted: 08/06/2021] [Indexed: 12/24/2022] Open
Abstract
Circadian clocks are ubiquitous timing mechanisms that generate approximately 24-h rhythms in cellular and bodily functions across nearly all living species. These internal clock systems enable living organisms to anticipate and respond to daily changes in their environment in a timely manner, optimizing temporal physiology and behaviors. Dysregulation of circadian rhythms by genetic and environmental risk factors increases susceptibility to multiple diseases, particularly cancers. A growing number of studies have revealed dynamic crosstalk between circadian clocks and cancer pathways, providing mechanistic insights into the therapeutic utility of circadian rhythms in cancer treatment. This review will discuss the roles of circadian rhythms in cancer pathogenesis, highlighting the recent advances in chronotherapeutic approaches for improved cancer treatment.
Collapse
Affiliation(s)
- Yool Lee
- grid.30064.310000 0001 2157 6568Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202 USA
| |
Collapse
|
16
|
Sato A, Rahman NIA, Shimizu A, Ogita H. Cell-to-cell contact-mediated regulation of tumor behavior in the tumor microenvironment. Cancer Sci 2021; 112:4005-4012. [PMID: 34420253 PMCID: PMC8486192 DOI: 10.1111/cas.15114] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023] Open
Abstract
Tumor growth and progression are complex processes mediated by mutual interactions between cancer cells and their surrounding stroma that include diverse cell types and acellular components, which form the tumor microenvironment. In this environment, direct intercellular communications play important roles in the regulation of the biological behaviors of tumors. However, the underlying molecular mechanisms are insufficiently defined. We used an in vitro coculture system to identify genes that were specifically expressed at higher levels in cancer cells associated with stromal cells. Major examples included epithelial membrane protein 1 (EMP1) and stomatin, which positively and negatively regulate tumor progression, respectively. EMP1 promotes tumor cell migration and metastasis via activation of the small GTPase Rac1, while stomatin strongly suppresses cell proliferation and induces apoptosis of cancer cells via inhibition of Akt signaling. Here we highlight important aspects of EMP1, stomatin, and their family members in cancer biology. Furthermore, we consider the molecules that participate in intercellular communications and signaling transduction between cancer cells and stromal cells, which may affect the phenotypes of cancer cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Akira Sato
- Division of Molecular Medical BiochemistryDepartment of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Nor Idayu A. Rahman
- Division of Molecular Medical BiochemistryDepartment of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Akio Shimizu
- Division of Molecular Medical BiochemistryDepartment of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Hisakazu Ogita
- Division of Molecular Medical BiochemistryDepartment of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| |
Collapse
|
17
|
Khan M, Li X, Yan M, Li Z, Yang H, Liao G. Efficacy and Safety of Actively Personalized Neoantigen Vaccination in the Management of Newly Diagnosed Glioblastoma: A Systematic Review. Int J Gen Med 2021; 14:5209-5220. [PMID: 34512004 PMCID: PMC8427683 DOI: 10.2147/ijgm.s323576] [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: 06/10/2021] [Accepted: 08/20/2021] [Indexed: 12/30/2022] Open
Abstract
Purpose Glioblastoma (GBM) shows frequent relapse and is highly resistant to treatment; therefore, it is considered fatal. Various vaccination protocols that have been tested in patients with GBM, which is the most common and aggressive primary brain tumor, have indicated safety and efficacy, to some extent, when used alone or in combination with standard of care. Recently, neoantigen-based personalized vaccines have shown tremendous immunogenicity and safety in GBM. We aimed to systematically review the medical literature for clinical trials to evaluate the efficacy and safety of neoantigen-based personalized vaccines for newly diagnosed GBM. Methods We conducted a literature search for clinical trials on PubMed, Cochrane Library, China National Knowledge Infrastructure, and ClinicalTrials.gov until March 20, 2021. The primary outcomes of interest were immunogenicity and safety of the therapy. Efficacy outcomes, such as progression-free survival and overall survival, were secondary outcomes of interest. Results Two clinical trials involving 24 patients were included in this review. High immunogenicity was observed in both studies. The GAPVAC-101 trial reported 50% APVAC1-induced and 84.7% APVAC2-induced immunogenicity with CD8+ and CD4+ T cell responses in 92% (12/13) and 80% (8/10) immune responders, respectively. Two out of five patients showed CD4+ and CD8+ T cell responses in the study by Keskin et al. Dexamethasone use had limited immunogenicity in a trial by Keskin et al (6/8). No serious treatment-related adverse events were reported. Conclusion Actively personalized vaccines aimed at unmutated peptides and neoantigens for patients with GBM are safe and highly immunogenic, particularly when administered in combination. Larger studies are warranted to investigate the role.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, People's Republic of China.,Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People's Republic of China
| | - Xianming Li
- Department of Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, People's Republic of China
| | - Maosheng Yan
- Department of Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, People's Republic of China
| | - Zihuang Li
- Department of Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, People's Republic of China
| | - Hongli Yang
- Department of Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, People's Republic of China
| | - Guixiang Liao
- Department of Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, People's Republic of China
| |
Collapse
|
18
|
Bange EM, Han NA, Wileyto P, Kim JY, Gouma S, Robinson J, Greenplate AR, Hwee MA, Porterfield F, Owoyemi O, Naik K, Zheng C, Galantino M, Weisman AR, Ittner CAG, Kugler EM, Baxter AE, Oniyide O, Agyekum RS, Dunn TG, Jones TK, Giannini HM, Weirick ME, McAllister CM, Babady NE, Kumar A, Widman AJ, DeWolf S, Boutemine SR, Roberts C, Budzik KR, Tollett S, Wright C, Perloff T, Sun L, Mathew D, Giles JR, Oldridge DA, Wu JE, Alanio C, Adamski S, Garfall AL, Vella LA, Kerr SJ, Cohen JV, Oyer RA, Massa R, Maillard IP, Maxwell KN, Reilly JP, Maslak PG, Vonderheide RH, Wolchok JD, Hensley SE, Wherry EJ, Meyer NJ, DeMichele AM, Vardhana SA, Mamtani R, Huang AC. CD8 + T cells contribute to survival in patients with COVID-19 and hematologic cancer. Nat Med 2021; 27:1280-1289. [PMID: 34017137 PMCID: PMC8291091 DOI: 10.1038/s41591-021-01386-7] [Citation(s) in RCA: 337] [Impact Index Per Article: 112.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
Patients with cancer have high mortality from coronavirus disease 2019 (COVID-19), and the immune parameters that dictate clinical outcomes remain unknown. In a cohort of 100 patients with cancer who were hospitalized for COVID-19, patients with hematologic cancer had higher mortality relative to patients with solid cancer. In two additional cohorts, flow cytometric and serologic analyses demonstrated that patients with solid cancer and patients without cancer had a similar immune phenotype during acute COVID-19, whereas patients with hematologic cancer had impairment of B cells and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific antibody responses. Despite the impaired humoral immunity and high mortality in patients with hematologic cancer who also have COVID-19, those with a greater number of CD8 T cells had improved survival, including those treated with anti-CD20 therapy. Furthermore, 77% of patients with hematologic cancer had detectable SARS-CoV-2-specific T cell responses. Thus, CD8 T cells might influence recovery from COVID-19 when humoral immunity is deficient. These observations suggest that CD8 T cell responses to vaccination might provide protection in patients with hematologic cancer even in the setting of limited humoral responses.
Collapse
Affiliation(s)
- Erin M Bange
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas A Han
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Wileyto
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Justin Y Kim
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James Robinson
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Allison R Greenplate
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Madeline A Hwee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Florence Porterfield
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olutosin Owoyemi
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karan Naik
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cathy Zheng
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Galantino
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ariel R Weisman
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline A G Ittner
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily M Kugler
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy E Baxter
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olutwatosin Oniyide
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital, Philadelphia, PA, USA
| | - Roseline S Agyekum
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital, Philadelphia, PA, USA
| | - Thomas G Dunn
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital, Philadelphia, PA, USA
| | - Tiffanie K Jones
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital, Philadelphia, PA, USA
| | - Heather M Giannini
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Madison E Weirick
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher M McAllister
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - N Esther Babady
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anita Kumar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adam J Widman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Susan DeWolf
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sawsan R Boutemine
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlotte Roberts
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Krista R Budzik
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan Tollett
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Carla Wright
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Tara Perloff
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital, Philadelphia, NY, USA
| | - Lova Sun
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Josephine R Giles
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Derek A Oldridge
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer E Wu
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Cécile Alanio
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Sharon Adamski
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alfred L Garfall
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura A Vella
- Department of Pediatrics, Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Samuel J Kerr
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital, Philadelphia, PA, USA
| | - Justine V Cohen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital, Philadelphia, NY, USA
| | - Randall A Oyer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital, Philadelphia, PA, USA
| | - Ryan Massa
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital, Philadelphia, PA, USA
| | - Ivan P Maillard
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Kara N Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - John P Reilly
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter G Maslak
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert H Vonderheide
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Jedd D Wolchok
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Scott E Hensley
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - E John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
| | - Nuala J Meyer
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angela M DeMichele
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Santosha A Vardhana
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA.
| | - Ronac Mamtani
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| | - Alexander C Huang
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Nayak SP, Roy S. Immune phase transition under steroid treatment. Phys Rev E 2021; 103:062401. [PMID: 34271610 DOI: 10.1103/physreve.103.062401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/11/2021] [Indexed: 11/07/2022]
Abstract
The steroid hormone glucocorticoid (GC) is a well-known immunosuppressant that controls T-cell-mediated adaptive immune response. In this work, we have developed a minimal kinetic network model of T-cell regulation connecting relevant experimental and clinical studies to quantitatively understand the long-term effects of GC on pro-inflammatory T-cell (T_{pro}) and anti-inflammatory T-cell (T_{anti}) dynamics. Due to the antagonistic relation between these two types of T cells, their long-term steady-state population ratio helps us to characterize three classified immune regulations: (i) weak ([T_{pro}]>[T_{anti}]), (ii) strong ([T_{pro}]<[T_{anti}]), and (iii) moderate ([T_{pro}]∼[T_{anti}]), holding the characteristic bistability. In addition to the differences in their long-term steady-state outcome, each immune regulation shows distinct dynamical phases. In the presteady state, a characteristic intermediate stationary phase is observed to develop only in the moderate regulation regime. In the medicinal field, the resting time in this stationary phase is distinguished as a clinical latent period. GC dose-dependent steady-state analysis shows an optimal level of GC to drive a phase transition from the weak or autoimmune prone to the moderate regulation regime. Subsequently, the presteady state clinical latent period tends to diverge near that optimal GC level where [T_{pro}]:[T_{anti}] is highly balanced. The GC-optimized elongated stationary phase explains the rationale behind the requirement of long-term immune diagnostics, especially when long-term GC-based chemotherapeutics and other immunosuppressive drugs are administrated. Moreover, our study reveals GC sensitivity of clinical latent period, which might serve as an early warning signal in diagnosing different immune phases and determining immune phasewise steroid treatment.
Collapse
Affiliation(s)
| | - Susmita Roy
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Campus Road, Mohanpur, West Bengal 741246, India
| |
Collapse
|
20
|
Amere Subbarao S. Cancer vs. SARS-CoV-2 induced inflammation, overlapping functions, and pharmacological targeting. Inflammopharmacology 2021; 29:343-366. [PMID: 33723711 PMCID: PMC7959277 DOI: 10.1007/s10787-021-00796-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/27/2021] [Indexed: 12/15/2022]
Abstract
Inflammation is an intrinsic defence mechanism triggered by the immune system against infection or injury. Chronic inflammation allows the host to recover or adapt through cellular and humoral responses, whereas acute inflammation leads to cytokine storms resulting in tissue damage. In this review, we present the overlapping outcomes of cancer inflammation with virus-induced inflammation. The study emphasises how anti-inflammatory drugs that work against cancer inflammation may work against the inflammation caused by the viral infection. It is established that the cytokine storm induced in response to SARS-CoV-2 infection contributes to disease-associated mortality. While cancer remains the second among the diseases associated with mortality worldwide, cancer patients' mortality rates are often observed upon extended periods after illness, usually ranging from months to years. However, the mortality rates associated with COVID-19 disease are robust. The cytokine storm induced by SARS-CoV-2 infection appeared to be responsible for the multi-organ failure and increased mortality rates. Since both cancer and COVID-19 disease share overlapping inflammatory mechanisms, repurposing some anticancer and anti-inflammatory drugs for COVID-19 may lower mortality rates. Here, we review some of these inflammatory mechanisms and propose some potential chemotherapeutic agents to intervene in them. We also discuss the repercussions of anti-inflammatory drugs such as glucocorticoids and hydroxychloroquine with zinc or antiviral drugs such as ivermectin and remdesivir against SARS-CoV-2 induced cytokine storm. In this review, we emphasise on various possibilities to reduce SARS-CoV-2 induced cytokine storm.
Collapse
|
21
|
Rahman NIA, Sato A, Tsevelnorov K, Shimizu A, Komeno M, Ahmat Amin MKB, Molla MR, Soh JEC, Nguyen LKC, Wada A, Kawauchi A, Ogita H. Stomatin-Mediated Inhibition of the Akt Signaling Axis Suppresses Tumor Growth. Cancer Res 2021; 81:2318-2331. [PMID: 33757977 DOI: 10.1158/0008-5472.can-20-2331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 02/16/2021] [Accepted: 03/11/2021] [Indexed: 11/16/2022]
Abstract
The growth and progression of cancers are crucially regulated by the tumor microenvironment where tumor cells and stromal cells are mutually associated. In this study, we found that stomatin expression was markedly upregulated by the interaction between prostate cancer cells and stromal cells. Stomatin suppressed cancer cell proliferation and enhanced apoptosis in vitro and inhibited xenograft tumor growth in vivo. Stomatin inhibited Akt activation, which is mediated by phosphoinositide-dependent protein kinase 1 (PDPK1). PDPK1 protein stability was maintained by its binding to HSP90. Stomatin interacted with PDPK1 and interfered with the PDPK1-HSP90 complex formation, resulting in decreased PDPK1 expression. Knockdown of stomatin in cancer cells elevated Akt activation and promoted cell increase by promoting the interaction between PDPK1 and HSP90. Clinically, stomatin expression levels were significantly decreased in human prostate cancer samples with high Gleason scores, and lower expression of stomatin was associated with higher recurrence of prostate cancer after the operation. Collectively, these findings demonstrate the tumor-suppressive effect of stromal-induced stomatin on cancer cells. SIGNIFICANCE: These findings reveal that interactions with stromal cells induce expression of stomatin in prostate cancer cells, which suppresses tumor growth via attenuation of the Akt signaling axis.
Collapse
Affiliation(s)
- Nor Idayu A Rahman
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan.
| | - Khurelbaatar Tsevelnorov
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Masahiro Komeno
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Mohammad Khusni Bin Ahmat Amin
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Md Rasel Molla
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Joanne Ern Chi Soh
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Le Kim Chi Nguyen
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akinori Wada
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - Akihiro Kawauchi
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan.
| |
Collapse
|
22
|
Cecchi M, Paccosi S, Silvano A, Eid AH, Parenti A. Dexamethasone Induces the Expression and Function of Tryptophan-2-3-Dioxygenase in SK-MEL-28 Melanoma Cells. Pharmaceuticals (Basel) 2021; 14:ph14030211. [PMID: 33806305 PMCID: PMC7998133 DOI: 10.3390/ph14030211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/26/2021] [Accepted: 02/28/2021] [Indexed: 12/18/2022] Open
Abstract
Tryptophan-2,3-dioxygenase (TDO) is one of the key tryptophan-catabolizing enzymes with immunoregulatory properties in cancer. Contrary to expectation, clinical trials showed that inhibitors of the ubiquitously expressed enzyme, indoleamine-2,3-dioxygenase-1 (IDO1), do not provide benefits in melanoma patients. This prompted the hypothesis that TDO may be a more attractive target. Because the promoter of TDO harbors glucocorticoid response elements (GREs), we aimed to assess whether dexamethasone (dex), a commonly used glucocorticoid, modulates TDO expression by means of RT-PCR and immunofluorescence and function by assessing cell proliferation and migration as well as metalloproteinase activity. Our results show that, in SK-Mel-28 melanoma cells, dex up-regulated TDO and its downstream effector aryl hydrocarbon receptor (AHR) but not IDO1. Furthermore, dex stimulated cellular proliferation and migration and potentiated MMP2 activity. These effects were inhibited by the selective TDO inhibitor 680C91 and enhanced by IDO1 inhibitors. Taken together, our results demonstrate that the metastatic melanoma cell line SK-Mel-28 possesses a functional TDO which can also modulate cancer cell phenotype directly rather than through immune suppression. Thus, TDO appears to be a promising, tractable target in the management or the treatment of melanoma progression.
Collapse
Affiliation(s)
- Marta Cecchi
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (M.C.); (S.P.); (A.S.)
| | - Sara Paccosi
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (M.C.); (S.P.); (A.S.)
| | - Angela Silvano
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (M.C.); (S.P.); (A.S.)
| | - Ali Hussein Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
- Correspondence: (A.H.E.); (A.P.); Tel.: +974-4403-7893 (A.H.E.)
| | - Astrid Parenti
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (M.C.); (S.P.); (A.S.)
- Correspondence: (A.H.E.); (A.P.); Tel.: +974-4403-7893 (A.H.E.)
| |
Collapse
|
23
|
Bange EM, Han NA, Wileyto P, Kim JY, Gouma S, Robinson J, Greenplate AR, Porterfield F, Owoyemi O, Naik K, Zheng C, Galantino M, Weisman AR, Ittner CA, Kugler EM, Baxter AE, Oniyide O, Agyekum RS, Dunn TG, Jones TK, Giannini HM, Weirick ME, McAllister CM, Babady NE, Kumar A, Widman AJ, DeWolf S, Boutemine SR, Roberts C, Budzik KR, Tollett S, Wright C, Perloff T, Sun L, Mathew D, Giles JR, Oldridge DA, Wu JE, Alanio C, Adamski S, Garfall AL, Vella L, Kerr SJ, Cohen JV, Oyer RA, Massa R, Maillard IP, Maxwell KN, Reilly JP, Maslak PG, Vonderheide RH, Wolchok JD, Hensley SE, Wherry EJ, Meyer N, DeMichele AM, Vardhana SA, Mamtani R, Huang AC. CD8 T cells compensate for impaired humoral immunity in COVID-19 patients with hematologic cancer. RESEARCH SQUARE 2021:rs.3.rs-162289. [PMID: 33564756 PMCID: PMC7872363 DOI: 10.21203/rs.3.rs-162289/v1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer patients have increased morbidity and mortality from Coronavirus Disease 2019 (COVID-19), but the underlying immune mechanisms are unknown. In a cohort of 100 cancer patients hospitalized for COVID-19 at the University of Pennsylvania Health System, we found that patients with hematologic cancers had a significantly higher mortality relative to patients with solid cancers after accounting for confounders including ECOG performance status and active cancer status. We performed flow cytometric and serologic analyses of 106 cancer patients and 113 non-cancer controls from two additional cohorts at Penn and Memorial Sloan Kettering Cancer Center. Patients with solid cancers exhibited an immune phenotype similar to non-cancer patients during acute COVID-19 whereas patients with hematologic cancers had significant impairment of B cells and SARS-CoV-2-specific antibody responses. High dimensional analysis of flow cytometric data revealed 5 distinct immune phenotypes. An immune phenotype characterized by CD8 T cell depletion was associated with a high viral load and the highest mortality of 71%, among all cancer patients. In contrast, despite impaired B cell responses, patients with hematologic cancers and preserved CD8 T cells had a lower viral load and mortality. These data highlight the importance of CD8 T cells in acute COVID-19, particularly in the setting of impaired humoral immunity. Further, depletion of B cells with anti-CD20 therapy resulted in almost complete abrogation of SARS-CoV-2-specific IgG and IgM antibodies, but was not associated with increased mortality compared to other hematologic cancers, when adequate CD8 T cells were present. Finally, higher CD8 T cell counts were associated with improved overall survival in patients with hematologic cancers. Thus, CD8 T cells likely compensate for deficient humoral immunity and influence clinical recovery of COVID-19. These observations have important implications for cancer and COVID-19-directed treatments, immunosuppressive therapies, and for understanding the role of B and T cells in acute COVID-19.
Collapse
Affiliation(s)
- Erin M. Bange
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Nicholas A. Han
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
| | - Paul Wileyto
- Abramson Cancer Center, University of Pennsylvania
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania
| | - Justin Y. Kim
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | | | - Allison R. Greenplate
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Florence Porterfield
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Olutosin Owoyemi
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Karan Naik
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Cathy Zheng
- Abramson Cancer Center, University of Pennsylvania
| | | | - Ariel R. Weisman
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Caroline A.G. Ittner
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Emily M. Kugler
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Amy E. Baxter
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Olutwatosin Oniyide
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Roseline S. Agyekum
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Thomas G. Dunn
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Tiffanie K. Jones
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Heather M. Giannini
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Madison E. Weirick
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | | | - N. Esther Babady
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| | - Anita Kumar
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Adam J Widman
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Susan DeWolf
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | | | | | | | | | - Carla Wright
- Abramson Cancer Center, University of Pennsylvania
| | - Tara Perloff
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital
| | - Lova Sun
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Divij Mathew
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Josephine R. Giles
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Derek A. Oldridge
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Jennifer E. Wu
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Cécile Alanio
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Sharon Adamski
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
| | - Alfred L. Garfall
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Laura Vella
- Department of Pediatrics, Perelman School of Medicine, Children’s Hospital of Philadelphia
| | - Samuel J. Kerr
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital
| | - Justine V. Cohen
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Pennsylvania Hospital
| | - Randall A. Oyer
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Lancaster General Hospital
| | - Ryan Massa
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, Presbyterian Hospital
| | - Ivan P. Maillard
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | | | - Kara N. Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - John P. Reilly
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Peter G. Maslak
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| | - Robert H. Vonderheide
- Abramson Cancer Center, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Jedd D. Wolchok
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Scott E. Hensley
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania
| | - E. John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| | - Nuala Meyer
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Division of Pulmonary and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Angela M. DeMichele
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Santosha A. Vardhana
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Parker Institute for Cancer Immunotherapy
| | - Ronac Mamtani
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
| | - Alexander C. Huang
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania
- Abramson Cancer Center, University of Pennsylvania
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
- Parker Institute for Cancer Immunotherapy
| |
Collapse
|
24
|
Pathania AS, Prathipati P, Abdul BAA, Chava S, Katta SS, Gupta SC, Gangula PR, Pandey MK, Durden DL, Byrareddy SN, Challagundla KB. COVID-19 and Cancer Comorbidity: Therapeutic Opportunities and Challenges. Theranostics 2021; 11:731-753. [PMID: 33391502 PMCID: PMC7738845 DOI: 10.7150/thno.51471] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/12/2020] [Indexed: 01/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) is a viral disease caused by a novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that affects the respiratory system of infected individuals. COVID-19 spreads between humans through respiratory droplets produced when an infected person coughs or sneezes. The COVID-19 outbreak originated in Wuhan, China at the end of 2019. As of 29 Sept 2020, over 235 countries, areas or territories across the globe reported a total of 33,441,919 confirmed cases, and 1,003,497 confirmed deaths due to COVID-19. Individuals of all ages are at risk for infection, but in most cases disease severity is associated with age and pre-existing diseases that compromise immunity, like cancer. Numerous reports suggest that people with cancer can be at higher risk of severe illness and related deaths from COVID-19. Therefore, managing cancer care under this pandemic is challenging and requires a collaborative multidisciplinary approach for optimal care of cancer patients in hospital settings. In this comprehensive review, we discuss the impact of the COVID-19 pandemic on cancer patients, their care, and treatment. Further, this review covers the SARS-CoV-2 pandemic, genome characterization, COVID-19 pathophysiology, and associated signaling pathways in cancer, and the choice of anticancer agents as repurposed drugs for treating COVID-19.
Collapse
Affiliation(s)
- Anup S. Pathania
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center; University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Philip Prathipati
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition, Saito-Asagi Ibaraki City, Osaka 567-0085, Japan
| | - Bakrudeen AA. Abdul
- Department of Biochemistry, Center for Research & Development, PRIST Deemed University, Vallam, Tamil Nadu 613403, India
| | - Srinivas Chava
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center; University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Santharam S. Katta
- Department of Biotechnology, School of Applied Sciences, REVA University, Rukmini Knowledge Park Kattigenahalli, Yelahanka, Bangalore, Karnataka 560064, India
| | - Subash C. Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Pandu R. Gangula
- Department of Oral Diagnostic Sciences and Research, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA
| | - Manoj K. Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Donald L. Durden
- Levine Cancer Institute, Atrium Health, Charlotte, NC 28202, USA
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
- SignalRx Pharmaceuticals, Omaha, NE 68124, USA
| | - Siddappa N. Byrareddy
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center; University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kishore B. Challagundla
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center; University of Nebraska Medical Center, Omaha, NE 68198, USA
- The Children's Health Research Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
25
|
Kukkar D, Kukkar P, Kumar V, Hong J, Kim KH, Deep A. Recent advances in nanoscale materials for antibody-based cancer theranostics. Biosens Bioelectron 2020; 173:112787. [PMID: 33190049 DOI: 10.1016/j.bios.2020.112787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/08/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
The quest for advanced management tools or options of various cancers has been on the rise to efficiently reduce their risks of mortality without the demerits of conventional treatments (e.g., undesirable side effects of the medications on non-target tissues, non-targeted distribution, slow clearance of the administered drugs, and the development of drug resistance over the duration of therapy). In this context, nanomaterials-antibody conjugates can offer numerous advantages in the development of cancer theranostics over conventional delivery systems (e.g., highly specific and enhanced biodistribution of the drug in targeted tissues, prolonged systemic circulation, low toxicity, and minimally invasive molecular imaging). This review comprehensively discusses and evaluates recent advances in the application of nanomaterial-antibody bioconjugates for cancer theranostics for the further advancement in the control of diverse cancerous diseases. Further, discussion is expanded to cover the various challenges and limitations associated with the design and development of nanomaterial-antibody conjugates applicable towards better management of cancer.
Collapse
Affiliation(s)
- Deepak Kukkar
- Department of Nanotechnology, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, 140406, India
| | - Preeti Kukkar
- Department of Chemistry, Mata Gujri College, Fatehgarh Sahib, Punjab, 140406, India
| | - Vanish Kumar
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Punjab, 140306, India
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, Seoul, 04763 Republic of Korea.
| | - Akash Deep
- Central Scientific Instruments Organization (CSIR-CSIO), Sector 30 C, Chandigarh, 160030, India.
| |
Collapse
|
26
|
Alhmoud JF, Mustafa AG, Malki MI. Targeting DNA Repair Pathways in Hematological Malignancies. Int J Mol Sci 2020; 21:ijms21197365. [PMID: 33036137 PMCID: PMC7582413 DOI: 10.3390/ijms21197365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/25/2020] [Accepted: 10/01/2020] [Indexed: 12/19/2022] Open
Abstract
DNA repair plays an essential role in protecting cells that are repeatedly exposed to endogenous or exogenous insults that can induce varying degrees of DNA damage. Any defect in DNA repair mechanisms results in multiple genomic changes that ultimately may result in mutation, tumor growth, and/or cell apoptosis. Furthermore, impaired repair mechanisms can also lead to genomic instability, which can initiate tumorigenesis and development of hematological malignancy. This review discusses recent findings and highlights the importance of DNA repair components and the impact of their aberrations on hematological malignancies.
Collapse
Affiliation(s)
- Jehad F. Alhmoud
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan;
| | - Ayman G. Mustafa
- College of Medicine, QU Health, Qatar University, P. O. Box 2713 Doha, Qatar;
| | - Mohammed Imad Malki
- College of Medicine, QU Health, Qatar University, P. O. Box 2713 Doha, Qatar;
- Correspondence: ; Tel.: +97-44403-7847
| |
Collapse
|
27
|
Hesketh PJ, Kris MG, Basch E, Bohlke K, Barbour SY, Clark-Snow RA, Danso MA, Dennis K, Dupuis LL, Dusetzina SB, Eng C, Feyer PC, Jordan K, Noonan K, Sparacio D, Lyman GH. Antiemetics: ASCO Guideline Update. J Clin Oncol 2020; 38:2782-2797. [DOI: 10.1200/jco.20.01296] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE To update the guideline to include new anticancer agents, antiemetics, and antiemetic regimens and to provide recommendations on the use of dexamethasone as a prophylactic antiemetic in patients receiving checkpoint inhibitors (CPIs). METHODS ASCO convened an Expert Panel and updated the systematic review to include randomized controlled trials (RCTs) and meta-analyses of RCTs published between June 1, 2016, and January 24, 2020. To address the dexamethasone and CPI question, we conducted a systematic review of RCTs that evaluated the addition of a CPI to chemotherapy. RESULTS The systematic reviews included 3 publications from the updated search and 10 publications on CPIs. Two phase III trials in adult patients with non–small-cell lung cancers evaluating a platinum-based doublet with or without the programmed death 1 (PD-1) inhibitor pembrolizumab recommended that all patients receive dexamethasone as a component of the prophylactic antiemetic regimen. In both studies, superior outcomes were noted in the PD-1 inhibitor–containing arms. Other important findings address olanzapine in adults and fosaprepitant in pediatric patients. RECOMMENDATIONS Recommendations for adults are unchanged with the exception of the option of adding olanzapine in the setting of hematopoietic stem cell transplantation. Dosing information now includes the option of a 5-mg dose of olanzapine in adults and intravenous formulations of aprepitant and netupitant-palonosetron. The option of fosaprepitant is added to pediatric recommendations. There is no clinical evidence to warrant omission of dexamethasone from guideline-compliant prophylactic antiemetic regimens when CPIs are administered to adults in combination with chemotherapy. CPIs administered alone or in combination with another CPI do not require the routine use of a prophylactic antiemetic. Additional information is available at www.asco.org/supportive-care-guidelines .
Collapse
Affiliation(s)
| | - Mark G. Kris
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ethan Basch
- University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Kari Bohlke
- American Society of Clinical Oncology, Alexandria, VA
| | | | | | | | - Kristopher Dennis
- The Ottawa Hospital, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | - L. Lee Dupuis
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Stacie B. Dusetzina
- Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Cathy Eng
- Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Petra C. Feyer
- Clinic of Radio-Oncology and Nuclear Medicine, Vivantes Clinics Neukoelln, Berlin, Germany
| | - Karin Jordan
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | | - Gary H. Lyman
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA
| |
Collapse
|
28
|
DMBA-Induced Oral Carcinoma in Syrian Hamster: Increased Carcinogenic Effect by Dexamethasone Coexposition. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1470868. [PMID: 32149076 PMCID: PMC7042540 DOI: 10.1155/2020/1470868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/06/2019] [Accepted: 01/09/2020] [Indexed: 12/30/2022]
Abstract
Objectives To investigate the effect of systemic administration of the immunosuppressant dexamethasone (DM) while inducing hamster buccal pouch DMBA carcinogenesis. Materials and Methods. Two different experiments were performed. In the first experiment, hamsters' right buccal pouches in group A (n = 10) were painted three times per week with 7,12-dimethylbenzanthracene (DMBA) 0.5%, while pouches of animals in group B (n = 10) were painted three times per week with 7,12-dimethylbenzanthracene (DMBA) 0.5%, while pouches of animals in group B ( Results The time of macroscopic neoplasm development was reduced when DM-DMBA coexposition was employed, finding tumors after 10–12 weeks of exposition. In addition, the frequency of histopathological lesions was higher. Conclusion Immunomodulatory action of dexamethasone may reduce the time of oral squamous cell carcinoma (OSCC) induction and may increase the incidence of neoplasms developed.
Collapse
|
29
|
Cheng AL, Hsu C, Chan SL, Choo SP, Kudo M. Challenges of combination therapy with immune checkpoint inhibitors for hepatocellular carcinoma. J Hepatol 2020; 72:307-319. [PMID: 31954494 DOI: 10.1016/j.jhep.2019.09.025] [Citation(s) in RCA: 319] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/09/2019] [Accepted: 09/28/2019] [Indexed: 02/06/2023]
Abstract
Immune checkpoint inhibitor (ICI) therapy targeting anti-programmed cell death-1 (anti-PD-1) or its ligand (anti-PD-L1) is the backbone of numerous combination regimens aimed at improving the objective response and survival of patients with hepatocellular carcinoma (HCC). Clinical trials of immuno-oncology regimens in other cancer types have shed light on issues of study design, including how to choose candidate regimens based on early-phase trial results, statistical considerations in trials with multiple primary endpoints, and the importance of predictive biomarkers. In this review, the updated data from early-phase trials of combination immunotherapy for HCC are summarised. Since the most extensively tested combination regimens for advanced HCC comprise anti-PD-1/anti-PD-L1 agents plus antiangiogenic agents, the relative benefit and antitumor mechanism of antiangiogenic multikinase inhibitors versus specific VEGF/VEGFR inhibitors are discussed. Other critical issues in the development of combination immunotherapy, including optimal management of immune-related adverse events and the value of ICI therapy in combination with locoregional treatment for HCC, are also explored.
Collapse
Affiliation(s)
- Ann-Lii Cheng
- National Taiwan University Cancer Center, Taipei, Taiwan; Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chiun Hsu
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan; Center of Precision Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Stephen L Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Institute of Digestive Disease, The Chinese University of Hong Kong, China
| | | | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
30
|
Boudewijns S, Bloemendal M, de Haas N, Westdorp H, Bol KF, Schreibelt G, Aarntzen EHJG, Lesterhuis WJ, Gorris MAJ, Croockewit A, van der Woude LL, van Rossum MM, Welzen M, de Goede A, Hato SV, van der Graaf WTA, Punt CJA, Koornstra RHT, Gerritsen WR, Figdor CG, de Vries IJM. Autologous monocyte-derived DC vaccination combined with cisplatin in stage III and IV melanoma patients: a prospective, randomized phase 2 trial. Cancer Immunol Immunother 2020; 69:477-488. [PMID: 31980913 PMCID: PMC7044256 DOI: 10.1007/s00262-019-02466-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/28/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Autologous dendritic cell (DC) vaccines can induce tumor-specific T cells, but their effect can be counteracted by immunosuppressive mechanisms. Cisplatin has shown immunomodulatory effects in vivo which may enhance efficacy of DC vaccination. METHODS This is a prospective, randomized, open-label phase 2 study (NCT02285413) including stage III and IV melanoma patients receiving 3 biweekly vaccinations of gp100 and tyrosinase mRNA-loaded monocyte-derived DCs with or without cisplatin. Primary objectives were to study immunogenicity and feasibility, and secondary objectives were to assess toxicity and survival. RESULTS Twenty-two stage III and 32 stage IV melanoma patients were analyzed. Antigen-specific CD8+ T cells were found in 44% versus 67% and functional T cell responses in 28% versus 19% of skin-test infiltrating lymphocytes in patients receiving DC vaccination with and without cisplatin, respectively. Four patients stopped cisplatin because of toxicity and continued DC monotherapy. No therapy-related grade 3 or 4 adverse events occurred due to DC monotherapy. During combination therapy, one therapy-related grade 3 adverse event, decompensated heart failure due to fluid overload, occurred. The clinical outcome parameters did not clearly suggest significant differences. CONCLUSIONS Combination of DC vaccination and cisplatin in melanoma patients is feasible and safe, but does not seem to result in more tumor-specific T cell responses or improved clinical outcome, when compared to DC vaccination monotherapy.
Collapse
Affiliation(s)
- Steve Boudewijns
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Martine Bloemendal
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Nienke de Haas
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Pharmacy, Radboud University Medical center, Nijmegen, The Netherlands
| | - Harm Westdorp
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Kalijn F Bol
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Erik H J G Aarntzen
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W Joost Lesterhuis
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,School of Biomedical Sciences, University of Western Australia, Crawley, Australia
| | - Mark A J Gorris
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Alexandra Croockewit
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lieke L van der Woude
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michelle M van Rossum
- Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marieke Welzen
- Department of Pharmacy, Radboud University Medical center, Nijmegen, The Netherlands
| | - Anna de Goede
- Department of Pharmacy, Radboud University Medical center, Nijmegen, The Netherlands
| | - Stanleyson V Hato
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | - Cornelis J A Punt
- Department of Medical Oncology, Academic University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Rutger H T Koornstra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.,Oncological Center, Rijnstate Hospital, Arnhem, The Netherlands
| | - Winald R Gerritsen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands. .,Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
31
|
Sneddon S, Rive CM, Ma S, Dick IM, Allcock RJN, Brown SD, Holt RA, Watson M, Leary S, Lee YCG, Robinson BWS, Creaney J. Identification of a CD8+ T-cell response to a predicted neoantigen in malignant mesothelioma. Oncoimmunology 2019; 9:1684713. [PMID: 32002298 PMCID: PMC6959430 DOI: 10.1080/2162402x.2019.1684713] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 12/29/2022] Open
Abstract
Neoantigens present unique and specific targets for personalized cancer immunotherapy strategies. Given the low mutational burden yet immunotherapy responsiveness of malignant mesothelioma (MM) when compared to other carcinogen-induced malignancies, identifying candidate neoantigens and T cells that recognize them has been a challenge. We used pleural effusions to gain access to MM tumor cells as well as immune cells in order to characterize the tumor-immune interface in MM. We characterized the landscape of potential neoantigens from SNVs identified in 27 MM patients and performed whole transcriptome sequencing of cell populations from 18 patient-matched pleural effusions. IFNγ ELISpot was performed to detect a CD8+ T cell responses to predicted neoantigens in one patient. We detected a median of 68 (range 7–258) predicted neoantigens across the samples. Wild-type non-binding to mutant binding predicted neoantigens increased risk of death in a model adjusting for age, sex, smoking status, histology and treatment (HR: 33.22, CI: 2.55–433.02, p = .007). Gene expression analysis indicated a dynamic immune environment within the pleural effusions. TCR clonotypes increased with predicted neoantigen burden. A strong activated CD8+ T-cell response was identified for a predicted neoantigen produced by a spontaneous mutation in the ROBO3 gene. Despite the challenges associated with the identification of bonafide neoantigens, there is growing evidence that these molecular changes can provide an actionable target for personalized therapeutics in difficult to treat cancers. Our findings support the existence of candidate neoantigens in MM despite the low mutation burden of the tumor, and may present improved treatment opportunities for patients.
Collapse
Affiliation(s)
- Sophie Sneddon
- National Centre for Asbestos Related Disease, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Australia
| | - Craig M Rive
- National Centre for Asbestos Related Disease, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Australia
| | - Shaokang Ma
- National Centre for Asbestos Related Disease, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Australia
| | - Ian M Dick
- National Centre for Asbestos Related Disease, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Australia
| | - Richard J N Allcock
- Pathwest Laboratory Medicine, Western Australia, QEII Medical Centre, Nedlands, Australia.,School of Biomedical Sciences, University of Western Australia, Nedlands, Australia
| | - Scott D Brown
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Robert A Holt
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Mark Watson
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Australia
| | - Shay Leary
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Australia
| | - Y C Gary Lee
- National Centre for Asbestos Related Disease, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Australia.,Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, Australia
| | - Bruce W S Robinson
- National Centre for Asbestos Related Disease, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Australia.,Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, Australia
| | - Jenette Creaney
- National Centre for Asbestos Related Disease, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Australia
| |
Collapse
|
32
|
Flaherty RL, Falcinelli M, Flint MS. Stress and drug resistance in cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:773-786. [PMID: 35582576 PMCID: PMC8992509 DOI: 10.20517/cdr.2019.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/16/2019] [Accepted: 05/29/2019] [Indexed: 06/15/2023]
Abstract
Patients diagnosed with cancer often undergo considerable psychological distress, and the induction of the psychological stress response has been linked with a poor response to chemotherapy. The psychological stress response is mediated by fluctuations of the hormones glucocorticoids (GCs) and catecholamines. Binding to their respective receptors, GCs and the catecholamines adrenaline/noradrenaline are responsible for signalling a wide range of processes involved in cell survival, cell cycle and immune function. Synthetic GCs are also often prescribed as co-medication alongside chemotherapy, and increasing evidence suggests that GCs may induce chemoresistance in multiple cancer types. In this review, we bring together evidence linking psychological stress hormone signalling with resistance to chemo- and immune therapies, as well as mechanistic evidence regarding the effects of exogenous stress hormones on the efficacy of chemotherapies.
Collapse
Affiliation(s)
- Renée L. Flaherty
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Moulsecoomb, Brighton, BN2 4GJ, UK
| | - Marta Falcinelli
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Moulsecoomb, Brighton, BN2 4GJ, UK
| | - Melanie S. Flint
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Moulsecoomb, Brighton, BN2 4GJ, UK
| |
Collapse
|
33
|
Ide H, Inoue S, Mizushima T, Kashiwagi E, Zheng Y, Miyamoto H. Role of glucocorticoid signaling in urothelial tumorigenesis: Inhibition by prednisone presumably through inducing glucocorticoid receptor transrepression. Mol Carcinog 2019; 58:2297-2305. [PMID: 31535408 DOI: 10.1002/mc.23118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022]
Abstract
Glucocorticoids, including dexamethasone (DEX) and prednisone (PRED), have been prescribed in patients with neoplastic disease as cytotoxic agents or comedications. Nonetheless, it remains uncertain whether they have an impact on the development of bladder cancer. We, therefore, assessed the functional role of the glucocorticoid-mediated glucocorticoid receptor (GR) signaling in urothelial tumorigenesis. Tumor formation was significantly delayed in xenograft-bearing mice with implantation of control bladder cancer UMUC3 cells or nonneoplastic urothelial SVHUC cells undergoing malignant transformation induced by a chemical carcinogen 3-methylcholanthrene (MCA), compared with respective GR knockdown xenografts. Using the in vitro system with MCA-SVHUC cells, we screened 11 GR ligands, including DEX, and found significant inhibitory effects of PRED on their neoplastic transformation. The effects of PRED were restored by a GR antagonist RU486 in GR-positive MCA-SVHUC cells, while PRED failed to inhibit the neoplastic transformation of GR knockdown cells. Significant decreases in the expression levels of oncogenes (c-Fos/c-Jun) and significant increases in those of a tumor suppressor UGT1A were seen in MCA-SVHUC-control cells (vs GR-short hairpin RNA) or PRED-treated MCA-SVHUC-control cells (vs mock). In addition, N-butyl-N-(4-hydroxybutyl) nitrosamine induced bladder cancer in all of eight mock-treated mice vs seven (87.5%) of DEX-treated (P = .302) or four (50%) of PRED-treated (P = .021) animals. Finally, DEX was found to considerably induce both transactivation (activation of glucocorticoid-response element mediated transcription and expression of its targets) and transrepression (suppression of nuclear factor-kappa B transactivation and expression of its regulated genes) of GR in SVHUC cells, while PRED more selectively induced GR transrepression. These findings suggest that PRED could prevent urothelial tumorigenesis presumably via inducing GR transrepression.
Collapse
Affiliation(s)
- Hiroki Ide
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Satoshi Inoue
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York.,James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Taichi Mizushima
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York.,James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Eiji Kashiwagi
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yichun Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
| | - Hiroshi Miyamoto
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York.,James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York.,Department of Urology, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
34
|
Paudel S, Shrestha A, Cho P, Shrestha R, Kim Y, Lee T, Kim JH, Jeong TC, Lee ES, Lee S. Assessing Drug Interaction and Pharmacokinetics of Loxoprofen in Mice Treated with CYP3A Modulators. Pharmaceutics 2019; 11:pharmaceutics11090479. [PMID: 31527544 PMCID: PMC6781309 DOI: 10.3390/pharmaceutics11090479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 11/16/2022] Open
Abstract
Loxoprofen (LOX) is a non-selective cyclooxygenase inhibitor that is widely used for the treatment of pain and inflammation caused by chronic and transitory conditions. Its alcoholic metabolites are formed by carbonyl reductase (CR) and they consist of trans-LOX, which is active, and cis-LOX, which is inactive. In addition, LOX can also be converted into an inactive hydroxylated metabolite (OH-LOXs) by cytochrome P450 (CYP). In a previous study, we reported that CYP3A4 is primarily responsible for the formation of OH-LOX in human liver microsomes. Although metabolism by CYP3A4 does not produce active metabolites, it can affect the conversion of LOX into trans-/cis-LOX, since CYP3A4 activity modulates the substrate LOX concentration. Although the pharmacokinetics (PK) and metabolism of LOX have been well defined, its CYP-related interactions have not been fully characterized. Therefore, we investigated the metabolism of LOX after pretreatment with dexamethasone (DEX) and ketoconazole (KTC), which induce and inhibit the activities of CYP3A, respectively. We monitored their effects on the PK parameters of LOX, cis-LOX, and trans-LOX in mice, and demonstrated that their PK parameters significantly changed in the presence of DEX or KTC pretreatment. Specifically, DEX significantly decreased the concentration of the LOX active metabolite formed by CR, which corresponded to an increased concentration of OH-LOX formed by CYP3A4. The opposite result occurred with KTC (a CYP3A inhibitor) pretreatment. Thus, we conclude that concomitant use of LOX with CYP3A modulators may lead to drug–drug interactions and result in minor to severe toxicity even though there is no direct change in the metabolic pathway that forms the LOX active metabolite.
Collapse
Affiliation(s)
- Sanjita Paudel
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | | | - Piljoung Cho
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Riya Shrestha
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Younah Kim
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Taeho Lee
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Ju-Hyun Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Korea.
| | - Tae Cheon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Korea.
| | - Eung-Seok Lee
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Korea.
| | - Sangkyu Lee
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| |
Collapse
|
35
|
Dubinski D, Hattingen E, Senft C, Seifert V, Peters KG, Reiss Y, Devraj K, Plate KH. Controversial roles for dexamethasone in glioblastoma - Opportunities for novel vascular targeting therapies. J Cereb Blood Flow Metab 2019; 39:1460-1468. [PMID: 31238763 PMCID: PMC6681527 DOI: 10.1177/0271678x19859847] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Glioblastoma is a highly aggressive and treatment resistant primary brain tumor. Features of glioblastoma include peritumoral cerebral edema, the major contributor to neurological impairment. Although the current clinical approach to edema management is administration of the synthetic corticoid dexamethasone, increasing evidence indicates numerous adverse effects of dexamethasone on glioblastoma burden at the molecular, cellular and clinical level. The contradictions of dexamethasone for glioblastoma and brain metastasis therapy are discussed in this article. Finally, alternative strategies for cerebrovascular edema therapy with vascular stabilizing, anti-permeability agents that are either approved or in clinical trials for diabetic retinopathy and macula edema, are addressed.
Collapse
Affiliation(s)
- Daniel Dubinski
- 1 Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt am Main, Germany.,2 Department of Neurosurgery, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Elke Hattingen
- 3 Department of Neuroradiology, University Hospital, Goethe University, Frankfurt am Main, Germany.,4 Frankfurt Cancer Institute, Frankfurt, Germany
| | - Christian Senft
- 2 Department of Neurosurgery, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Volker Seifert
- 2 Department of Neurosurgery, University Hospital, Goethe University, Frankfurt am Main, Germany
| | | | - Yvonne Reiss
- 1 Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt am Main, Germany.,4 Frankfurt Cancer Institute, Frankfurt, Germany.,6 LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt am Main, Germany.,7 German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kavi Devraj
- 1 Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt am Main, Germany.,6 LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt am Main, Germany
| | - Karl H Plate
- 1 Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt am Main, Germany.,4 Frankfurt Cancer Institute, Frankfurt, Germany.,6 LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt am Main, Germany.,7 German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
36
|
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: 2.6] [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
|
37
|
Khayeka-Wandabwa C, Ma X, Cao X, Nunna V, Pathak JL, Bernhardt R, Cai P, Bureik M. Plasma membrane localization of CYP4Z1 and CYP19A1 and the detection of anti-CYP19A1 autoantibodies in humans. Int Immunopharmacol 2019; 73:64-71. [PMID: 31082724 DOI: 10.1016/j.intimp.2019.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/14/2019] [Accepted: 05/02/2019] [Indexed: 12/15/2022]
Abstract
It is thought that autoantibody (aAb) production can be caused by (aberrant) protein targeting to the plasma surface of cells. We recently demonstrated the presence of the human cytochrome P450 enzyme CYP4Z1 on the plasma membrane of MCF-7 breast cancer cells and the detection of high titers of anti-CYP4Z1 aAbs in breast cancer patients, but not in healthy controls. In the present study we show that cells of the normal breast cell line MCF-10A do not display CYP4Z1 on their surface. By contrast, we detected CYP19A1 (aromatase) on the plasma membrane of both cell lines. Interestingly, the presence of CYPs on the cell surface did not correlate with their relative expression levels in these cell lines. Indirect ELISA experiments demonstrated the presence of anti-CYP19A1 aAbs in female breast cancer patient sera as well as in male and female controls, respectively; aAb titers in all three groups varied considerably and overall, the results obtained for each group were not significantly different from those of either of the other two groups. Based on these data we propose the hypothesis that CYP translocation to the plasma membrane, but not the intracellular expression level, is the crucial precondition for the generation of anti-CYP aAbs.
Collapse
Affiliation(s)
- Christopher Khayeka-Wandabwa
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China
| | - Xiaoshuang Ma
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China
| | - Xiaolin Cao
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China
| | - Venkatrao Nunna
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China
| | - Janak L Pathak
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China
| | - Rita Bernhardt
- Institute of Biochemistry, Saarland University, Saarbruecken, Germany
| | - Pengcheng Cai
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Matthias Bureik
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China.
| |
Collapse
|
38
|
Cari L, De Rosa F, Nocentini G, Riccardi C. Context-Dependent Effect of Glucocorticoids on the Proliferation, Differentiation, and Apoptosis of Regulatory T Cells: A Review of the Empirical Evidence and Clinical Applications. Int J Mol Sci 2019; 20:E1142. [PMID: 30845709 PMCID: PMC6429178 DOI: 10.3390/ijms20051142] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/26/2019] [Accepted: 02/28/2019] [Indexed: 12/15/2022] Open
Abstract
Glucocorticoids (GCs) are widely used to treat several diseases because of their powerful anti-inflammatory and immunomodulatory effects on immune cells and non-lymphoid tissues. The effects of GCs on T cells are the most relevant in this regard. In this review, we analyze how GCs modulate the survival, maturation, and differentiation of regulatory T (Treg) cell subsets into both murine models and humans. In this way, GCs change the Treg cell number with an impact on the mid-term and long-term efficacy of GC treatment. In vitro studies suggest that the GC-dependent expansion of Treg cells is relevant when they are activated. In agreement with this observation, the GC treatment of patients with established autoimmune, allergic, or (auto)inflammatory diseases causes an expansion of Treg cells. An exception to this appears to be the local GC treatment of psoriatic lesions. Moreover, the effects on Treg number in patients with multiple sclerosis are uncertain. The effects of GCs on Treg cell number in healthy/diseased subjects treated with or exposed to allergens/antigens appear to be context-dependent. Considering the relevance of this effect in the maturation of the immune system (tolerogenic response to antigens), the success of vaccination (including desensitization), and the tolerance to xenografts, the findings must be considered when planning GC treatment.
Collapse
Affiliation(s)
- Luigi Cari
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia I-06129, Italy.
| | - Francesca De Rosa
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia I-06129, Italy.
| | - Giuseppe Nocentini
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia I-06129, Italy.
| | - Carlo Riccardi
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia I-06129, Italy.
| |
Collapse
|
39
|
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: 11.0] [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
|
40
|
Wu J, Waxman DJ. Immunogenic chemotherapy: Dose and schedule dependence and combination with immunotherapy. Cancer Lett 2019; 419:210-221. [PMID: 29414305 DOI: 10.1016/j.canlet.2018.01.050] [Citation(s) in RCA: 215] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/20/2022]
Abstract
Conventional cytotoxic cancer chemotherapy is often immunosuppressive and associated with drug resistance and tumor regrowth after a short period of tumor shrinkage or growth stasis. However, certain cytotoxic cancer chemotherapeutic drugs, including doxorubicin, mitoxantrone, and cyclophosphamide, can kill tumor cells by an immunogenic cell death pathway, which activates robust innate and adaptive anti-tumor immune responses and has the potential to greatly increase the efficacy of chemotherapy. Here, we review studies on chemotherapeutic drug-induced immunogenic cell death, focusing on how the choice of a conventional cytotoxic agent and its dose and schedule impact anti-tumor immune responses. We propose a strategy for effective immunogenic chemotherapy that employs a modified metronomic schedule for drug delivery, which we term medium-dose intermittent chemotherapy (MEDIC). Striking responses have been seen in preclinical cancer models using MEDIC, where an immunogenic cancer chemotherapeutic agent is administered intermittently and at an intermediate dose, designed to impart strong and repeated cytotoxic damage to tumors, and on a schedule compatible with activation of a sustained anti-tumor immune response, thereby maximizing anti-cancer activity. We also discuss strategies for combination chemo-immunotherapy, and we outline approaches to identify new immunogenic chemotherapeutic agents for drug development.
Collapse
Affiliation(s)
- Junjie Wu
- Department of Biology, Division of Cell and Molecular Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - David J Waxman
- Department of Biology, Division of Cell and Molecular Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA.
| |
Collapse
|
41
|
Stepanenko AA, Chekhonin VP. Recent Advances in Oncolytic Virotherapy and Immunotherapy for Glioblastoma: A Glimmer of Hope in the Search for an Effective Therapy? Cancers (Basel) 2018; 10:E492. [PMID: 30563098 PMCID: PMC6316815 DOI: 10.3390/cancers10120492] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/19/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023] Open
Abstract
To date, no targeted drugs, antibodies or combinations of chemotherapeutics have been demonstrated to be more efficient than temozolomide, or to increase efficacy of standard therapy (surgery, radiotherapy, temozolomide, steroid dexamethasone). According to recent phase III trials, standard therapy may ensure a median overall survival of up to 18⁻20 months for adult patients with newly diagnosed glioblastoma. These data explain a failure of positive non-controlled phase II trials to predict positive phase III trials and should result in revision of the landmark Stupp trial as a historical control for median overall survival in non-controlled trials. A high rate of failures in clinical trials and a lack of effective chemotherapy on the horizon fostered the development of conceptually distinct therapeutic approaches: dendritic cell/peptide immunotherapy, chimeric antigen receptor (CAR) T-cell therapy and oncolytic virotherapy. Recent early phase trials with the recombinant adenovirus DNX-2401 (Ad5-delta24-RGD), polio-rhinovirus chimera (PVSRIPO), parvovirus H-1 (ParvOryx), Toca 511 retroviral vector with 5-fluorocytosine, heat shock protein-peptide complex-96 (HSPPC-96) and dendritic cell vaccines, including DCVax-L vaccine, demonstrated that subsets of patients with glioblastoma/glioma may benefit from oncolytic virotherapy/immunotherapy (>3 years of survival after treatment). However, large controlled trials are required to prove efficacy of next-generation immunotherapeutics and oncolytic vectors.
Collapse
Affiliation(s)
- Aleksei A Stepanenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Kropotkinsky lane 23, 119034 Moscow, Russia.
| | - Vladimir P Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Kropotkinsky lane 23, 119034 Moscow, Russia.
- Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N. I. Pirogov Russian National Research Medical University, the Ministry of Health of the Russian Federation, Ostrovitianov str. 1, 117997 Moscow, Russia.
| |
Collapse
|
42
|
Meng G, Tang X, Yang Z, Zhao Y, Curtis JM, McMullen TPW, Brindley DN. Dexamethasone decreases the autotaxin-lysophosphatidate-inflammatory axis in adipose tissue: implications for the metabolic syndrome and breast cancer. FASEB J 2018; 33:1899-1910. [PMID: 30192654 DOI: 10.1096/fj.201801226r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Lysophosphatidate (LPA) signaling through 6 receptors is regulated by the balance of LPA production by autotaxin (ATX) vs. LPA degradation by lipid phosphate phosphatases (LPPs). LPA promotes an inflammatory cycle by increasing the synthesis of cyclooxygenase-2 and multiple inflammatory cytokines that stimulate further ATX production. We aimed to determine whether the anti-inflammatory glucocorticoid (GC) dexamethasone (Dex) functions partly by decreasing the ATX-LPA inflammatory cycle in adipose tissue, a major site of ATX secretion. Treatment of human adipose tissue with 10-1000 nM Dex decreased ATX secretion, increased LPP1 expression, and decreased mRNA expressions of IL-6, TNF-α, peroxisome proliferator-activated receptor (PPAR)-γ, and adiponectin. Cotreatment with rosiglitazone (an insulin sensitizer), insulin, or both abolished Dex-induced decreases in ATX and adiponectin secretion, but did not reverse Dex-induced decreases in secretions of 20 inflammatory cytokines and chemokines. Dex-treated mice exhibited lower ATX activity in plasma, brain, and adipose tissue; decreased mRNA levels for LPA and sphingosine 1-phosphate (S1P) receptors in brain; and decreased plasma concentrations of LPA and S1P. Our results establish a novel mechanism for the anti-inflammatory effects of Dex through decreased signaling by the ATX-LPA-inflammatory axis. The GC action in adipose tissue has implications for the pathogenesis of insulin resistance and obesity in metabolic syndrome and breast cancer treatment.-Meng, G., Tang, X., Yang, Z., Zhao, Y., Curtis, J. M., McMullen, T. P. W., Brindley, D. N. Dexamethasone decreases the autotaxin-lysophosphatidate-inflammatory axis in adipose tissue: implications for the metabolic syndrome and breast cancer.
Collapse
Affiliation(s)
- Guanmin Meng
- Signal Transduction Research Group, Department of Biochemistry, Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
| | - Xiaoyun Tang
- Signal Transduction Research Group, Department of Biochemistry, Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
| | - Zelei Yang
- Signal Transduction Research Group, Department of Biochemistry, Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
| | - YuanYuan Zhao
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada; and
| | - Jonathan M Curtis
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada; and
| | - Todd P W McMullen
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - David N Brindley
- Signal Transduction Research Group, Department of Biochemistry, Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
43
|
Maxwell R, Luksik AS, Garzon-Muvdi T, Hung AL, Kim ES, Wu A, Xia Y, Belcaid Z, Gorelick N, Choi J, Theodros D, Jackson CM, Mathios D, Ye X, Tran PT, Redmond KJ, Brem H, Pardoll DM, Kleinberg LR, Lim M. Contrasting impact of corticosteroids on anti-PD-1 immunotherapy efficacy for tumor histologies located within or outside the central nervous system. Oncoimmunology 2018; 7:e1500108. [PMID: 30524891 PMCID: PMC6279341 DOI: 10.1080/2162402x.2018.1500108] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 12/21/2022] Open
Abstract
Immune checkpoint blockade targeting programmed cell death protein 1 (PD-1) is emerging as an important treatment strategy in a growing list of cancers, yet its clinical benefits are limited to a subset of patients. Further investigation of tumor-intrinsic predictors of response and how extrinsic factors, such as iatrogenic immunosuppression caused by conventional therapies, impact the efficacy of anti-PD-1 therapy are paramount. Given the widespread use of corticosteroids in cancer management and their immunosuppressive nature, this study sought to determine how corticosteroids influence anti-PD-1 responses and whether their effects were dependent on tumor location within the periphery versus central nervous system (CNS), which may have a more limiting immune environment. In well-established anti-PD-1-responsive murine tumor models, corticosteroid therapy resulted in systemic immune effects, including severe and persistent reductions in peripheral CD4+ and CD8 + T cells. Corticosteroid treatment was found to diminish the efficacy of anti-PD-1 therapy in mice bearing peripheral tumors with responses correlating with peripheral CD8/Treg ratio changes. In contrast, in mice bearing intracranial tumors, corticosteroids did not abrogate the benefits conferred by anti-PD-1 therapy. Despite systemic immune changes, anti-PD-1-mediated antitumor immune responses remained intact during corticosteroid treatment in mice bearing intracranial tumors. These findings suggest that anti-PD-1 responses may be differentially impacted by concomitant corticosteroid use depending on tumor location within or outside the CNS. As an immune-specialized site, the CNS may potentially play a protective role against the immunosuppressive effects of corticosteroids, thus sustaining antitumor immune responses mediated by PD-1 blockade.
Collapse
Affiliation(s)
- Russell Maxwell
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA.,Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Hospital, Baltimore, USA
| | - Andrew S Luksik
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| | | | - Alice L Hung
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| | - Eileen S Kim
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| | - Adela Wu
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| | - Yuanxuan Xia
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| | - Zineb Belcaid
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| | - Noah Gorelick
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| | - John Choi
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| | - Debebe Theodros
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| | | | | | - Xiaobu Ye
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| | - Phuoc T Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Hospital, Baltimore, USA
| | - Kristin J Redmond
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Hospital, Baltimore, USA
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins Hospital, Baltimore, USA
| | - Lawrence R Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Hospital, Baltimore, USA
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, USA
| |
Collapse
|
44
|
Tallón de Lara P, Cecconi V, Hiltbrunner S, Yagita H, Friess M, Bode B, Opitz I, Vrugt B, Weder W, Stolzmann P, Felley-Bosco E, Stahel RA, Tischler V, Britschgi C, Soldini D, van den Broek M, Curioni-Fontecedro A. Gemcitabine Synergizes with Immune Checkpoint Inhibitors and Overcomes Resistance in a Preclinical Model and Mesothelioma Patients. Clin Cancer Res 2018; 24:6345-6354. [PMID: 30154226 DOI: 10.1158/1078-0432.ccr-18-1231] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/24/2018] [Accepted: 08/21/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Combination of immune checkpoint inhibitors with chemotherapy is under investigation for cancer treatment. EXPERIMENTAL DESIGN We studied the rationale of such a combination for treating mesothelioma, a disease with limited treatment options. RESULTS The combination of gemcitabine and immune checkpoint inhibitors outperformed immunotherapy alone with regard to tumor control and survival in a preclinical mesothelioma model; however, the addition of dexamethasone to gemcitabine and immune checkpoint inhibitors nullified the synergistic clinical response. Furthermore, treatment with gemcitabine plus anti-PD-1 resulted in an objective clinical response in two patients with mesothelioma, who were resistant to gemcitabine or anti-PD-1 as monotherapy. CONCLUSIONS Thus, treatment of mesothelioma with a combination of gemcitabine with immune checkpoint inhibitors is feasible and results in synergistic clinical response compared with single treatment in the absence of steroids.
Collapse
Affiliation(s)
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Martina Friess
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Beata Bode
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Bart Vrugt
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Walter Weder
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Paul Stolzmann
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | | | - Rolf A Stahel
- Department of Hematology and Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Verena Tischler
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Christian Britschgi
- Department of Hematology and Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Davide Soldini
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Maries van den Broek
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| | | |
Collapse
|
45
|
Lin CH, Chuang PY, You SL, Chiang CJ, Huang CS, Wang MY, Chao M, Lu YS, Cheng AL, Tang CH. Effect of glucocorticoid use on survival in patients with stage I-III breast cancer. Breast Cancer Res Treat 2018; 171:225-234. [PMID: 29761323 DOI: 10.1007/s10549-018-4787-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/09/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE Glucocorticoids (GCs) are commonly used in breast cancer patients to ameliorate emesis induced by chemotherapy. Some preclinical studies have suggested that systemic GCs might promote survival of estrogen receptor (ER)-negative breast cancer cells. This study aims to clarify their clinical effect on patient survival. METHODS A total of 18,596 women with newly diagnosed stage I-III breast cancer in 2002-2006 were identified from the Taiwan Cancer Database and drug treatment was examined from the Taiwan National Health Insurance Claims Database. Of these, 3989 who did not receive adjuvant chemotherapy (non-chemotherapy cohort) and 3237 patients who received six cycles of adjuvant anthracycline-based chemotherapy (anthracycline cohort) were included. The impact of GC use on survival was analyzed separately in these two cohorts using Cox proportional hazards models. RESULTS In the non-chemotherapy cohort, GC use was associated with aggressive clinicopathological features of breast cancer. High-dose GC was associated with shorter overall survival in univariate analysis but not in multivariate analysis. In the anthracycline cohort, multivariate analysis showed that GC use at each dose level was significantly associated with longer breast cancer-specific survival (HR 0.65, 0.70, and 0.70 for low-dose, median-dose, and high-dose GC, respectively) and overall survival (HR 0.72, 0.76, and 0.73, respectively) when compared with those receiving no GC. The associations were significant in both ER-positive and ER-negative subgroups for breast cancer-specific survival, and in ER-negative subgroup for overall survival. CONCLUSION Concomitant use of GC improved survival in patients receiving adjuvant anthracycline-based chemotherapy for stage I-III breast cancer.
Collapse
Affiliation(s)
- Ching-Hung Lin
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Oncology Center, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Po-Ya Chuang
- School of Health Care Administration, Taipei Medical University, No. 172-1, Keelung Road, Section 2, Taipei, 106, Taiwan
| | - San-Lin You
- Department of Public Health, College of Medicine, National Taiwan University, Taipei, Taiwan.,Big Data Research Centre, Fu-Jen Catholic University, New Taipei City, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Chun-Ju Chiang
- Taiwan Cancer Registry and Graduate Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chiun-Sheng Huang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Yang Wang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming Chao
- Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Yen-Shen Lu
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ann-Lii Cheng
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chao-Hsiun Tang
- School of Health Care Administration, Taipei Medical University, No. 172-1, Keelung Road, Section 2, Taipei, 106, Taiwan.
| |
Collapse
|
46
|
Bacac M, Colombetti S, Herter S, Sam J, Perro M, Chen S, Bianchi R, Richard M, Schoenle A, Nicolini V, Diggelmann S, Limani F, Schlenker R, Hüsser T, Richter W, Bray-French K, Hinton H, Giusti AM, Freimoser-Grundschober A, Lariviere L, Neumann C, Klein C, Umaña P. CD20-TCB with Obinutuzumab Pretreatment as Next-Generation Treatment of Hematologic Malignancies. Clin Cancer Res 2018; 24:4785-4797. [PMID: 29716920 DOI: 10.1158/1078-0432.ccr-18-0455] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/24/2018] [Accepted: 04/25/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Despite promising clinical activity, T-cell-engaging therapies including T-cell bispecific antibodies (TCB) are associated with severe side effects requiring the use of step-up-dosing (SUD) regimens to mitigate safety. Here, we present a next-generation CD20-targeting TCB (CD20-TCB) with significantly higher potency and a novel approach enabling safer administration of such potent drug.Experimental Design: We developed CD20-TCB based on the 2:1 TCB molecular format and characterized its activity preclinically. We also applied a single administration of obinutuzumab (Gazyva pretreatment, Gpt; Genentech/Roche) prior to the first infusion of CD20-TCB as a way to safely administer such a potent drug.Results: CD20-TCB is associated with a long half-life and high potency enabled by high-avidity bivalent binding to CD20 and head-to-tail orientation of B- and T-cell-binding domains in a 2:1 molecular format. CD20-TCB displays considerably higher potency than other CD20-TCB antibodies in clinical development and is efficacious on tumor cells expressing low levels of CD20. CD20-TCB also displays potent activity in primary tumor samples with low effector:target ratios. In vivo, CD20-TCB regresses established tumors of aggressive lymphoma models. Gpt enables profound B-cell depletion in peripheral blood and secondary lymphoid organs and reduces T-cell activation and cytokine release in the peripheral blood, thus increasing the safety of CD20-TCB administration. Gpt is more efficacious and safer than SUD.Conclusions: CD20-TCB and Gpt represent a potent and safer approach for treatment of lymphoma patients and are currently being evaluated in phase I, multicenter study in patients with relapsed/refractory non-Hodgkin lymphoma (NCT03075696). Clin Cancer Res; 24(19); 4785-97. ©2018 AACR See related commentary by Prakash and Diefenbach, p. 4631.
Collapse
Affiliation(s)
- Marina Bacac
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland.
| | - Sara Colombetti
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Sylvia Herter
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Johannes Sam
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Mario Perro
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Stanford Chen
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Roberta Bianchi
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Marine Richard
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Anne Schoenle
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Valeria Nicolini
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Sarah Diggelmann
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Florian Limani
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Ramona Schlenker
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Tamara Hüsser
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Wolfgang Richter
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, pRED, Basel, Switzerland
| | - Katharine Bray-French
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, pRED, Basel, Switzerland
| | - Heather Hinton
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Anna Maria Giusti
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Anne Freimoser-Grundschober
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Laurent Lariviere
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, pRED, Munich, Germany
| | - Christiane Neumann
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Pablo Umaña
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland.
| |
Collapse
|
47
|
Popilski H, Abtew E, Schwendeman S, Domb A, Stepensky D. Efficacy of paclitaxel/dexamethasone intra-tumoral delivery in treating orthotopic mouse breast cancer. J Control Release 2018; 279:1-7. [PMID: 29654797 DOI: 10.1016/j.jconrel.2018.04.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 03/30/2018] [Accepted: 04/06/2018] [Indexed: 12/12/2022]
Abstract
The effect of topical co-administration of promoter drugs with paclitaxel to increase anti-tumor effects of paclitaxel was investigated. Mice with orthotopic 4T1-Luc breast cancer received single intra-tumoral injection of a polymeric formulation with paclitaxel and a specific promoter drug. Several promoter drugs were evaluated, including: dexamethasone, losartan, nicotinamide, Azone, and oleic acid. Dexamethasone exhibited the highest effect on paclitaxel anti-tumor activity, in a dose-dependent fashion. However, this effect was accompanied by systemic effects of dexamethasone, and inability to prevent tumor metastasis to the lungs. Topical co-administration of promoter drugs with anti-cancer agents can enhance their anti-tumor effects. Further investigations are needed to identify the most efficient combinations of promoter and anti-cancer drugs, and their suitability for the clinical management of the breast cancer disease.
Collapse
Affiliation(s)
- Hen Popilski
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Esther Abtew
- Institute of Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Steven Schwendeman
- Department of Pharmaceutical Sciences and Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Abraham Domb
- Institute of Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - David Stepensky
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
48
|
Maurice-Dror C, Perets R, Bar-Sela G. Glucocorticoids as an adjunct to oncologic treatment in solid malignancies - Not an innocent bystander. Crit Rev Oncol Hematol 2018; 126:37-44. [PMID: 29759565 DOI: 10.1016/j.critrevonc.2018.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 01/02/2018] [Accepted: 03/21/2018] [Indexed: 11/30/2022] Open
Abstract
Glucocorticoids are steroidal hormones which exert their action via genomic and non-genomic mechanisms. In the clinical setting, glucocorticoids are utilized for their anti-inflammatory, anti-allergenic and immunomodulatory effects and for their well-established, pro-apoptotic effects on hematological malignancies. In the treatment of solid tumors, glucocorticoids serve primarily for alleviation of tumor- and treatment-related symptoms and in most cases are not considered to have a direct effect on tumor growth and spread. However, significant pre-clinical data suggest that glucocorticoids have diverse effects on tumor progression, both pro- and anti- tumorigenic. In contrast, the clinical data regarding the pro- and anti-tumorigenic effects of glucocorticoids on solid tumors is scarce, and summarized in this review. The following review presents the suggested glucocorticoids mechanism of action and the effects of glucocorticoids on tumor cells, on the tumor microenvironment and on tumor response to cytotoxic therapy, in the pre-clinical and clinical settings.
Collapse
Affiliation(s)
| | - Ruth Perets
- Division of Oncology, Rambam Health Care Campus, Israel; Technion-Israel Institute of Technology, Haifa, Israel
| | - Gil Bar-Sela
- Division of Oncology, Rambam Health Care Campus, Israel; Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
49
|
Chen Z, Zheng Y, Shi Y, Cui Z. Overcoming tumor cell chemoresistance using nanoparticles: lysosomes are beneficial for (stearoyl) gemcitabine-incorporated solid lipid nanoparticles. Int J Nanomedicine 2018; 13:319-336. [PMID: 29391792 PMCID: PMC5768424 DOI: 10.2147/ijn.s149196] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite recent advances in targeted therapies and immunotherapies, chemotherapy using cytotoxic agents remains an indispensable modality in cancer treatment. Recently, there has been a growing emphasis in using nanomedicine in cancer chemotherapy, and several nanomedicines have already been used clinically to treat cancers. There is evidence that formulating small molecular cancer chemotherapeutic agents into nanomedicines significantly modifies their pharmacokinetics and often improves their efficacy. Importantly, cancer cells often develop resistance to chemotherapy, and formulating anticancer drugs into nanomedicines also helps overcome chemoresistance. In this review, we briefly describe the different classes of cancer chemotherapeutic agents, their mechanisms of action and resistance, and evidence of overcoming the resistance using nanomedicines. We then emphasize on gemcitabine and our experience in discovering the unique (stearoyl) gemcitabine solid lipid nanoparticles that are effective against tumor cells resistant to gemcitabine and elucidate the underlying mechanisms. It seems that lysosomes, which are an obstacle in the delivery of many drugs, are actually beneficial for our (stearoyl) gemcitabine solid lipid nanoparticles to overcome tumor cell resistance to gemcitabine.
Collapse
Affiliation(s)
- Zhe Chen
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yuanqiang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yanchun Shi
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Zhengrong Cui
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.,Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
50
|
Glišić R, Čakić-Milošević M, Ukropina M. Immunohistochemical study of enteric nervous system in dexamethasone-treated rats. KRAGUJEVAC JOURNAL OF SCIENCE 2018. [DOI: 10.5937/kgjsci1840163g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|