1
|
A multicentre cohort study assessing the utility of routine blood tests as adjuncts to identify complete responders in rectal cancer following neoadjuvant chemoradiotherapy. Int J Colorectal Dis 2022; 37:957-965. [PMID: 35325271 PMCID: PMC8976819 DOI: 10.1007/s00384-022-04103-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE Management of rectal cancer with a complete clinical response (cCR) to neoadjuvant chemoradiotherapy (NACRT) is controversial. Some advocate "watch and wait" programmes and organ-preserving surgery. Central to these strategies is the ability to accurately preoperatively distinguish cCR from residual disease (RD). We sought to identify if post-NACRT (preoperative) inflammatory markers act as an adjunct to MRI and endoscopy findings for distinguishing cCR from RD in rectal cancer. METHODS Patients from three specialist rectal cancer centres were screened for inclusion (2010-2015). For inclusion, patients were required to have completed NACRT, had a post-NACRT MRI (to assess mrTRG) and proceeded to total mesorectal excision (TME). Endoluminal response was assessed on endoscopy at 6-8 weeks post-NACRT. Pathological response to therapy was calculated using a three-point tumour regression grade system (TRG1-3). Neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), serum albumin (SAL), CEA and CA19-9 levels post-NACRT (preoperatively) were recorded. Variables were compared between those who had RD on post-operative pathology and those with ypCR. Statistical analysis was performed using SPSS (version 21). RESULTS Six hundred forty-six patients were screened, of which 422 were suitable for inclusion. A cCR rate of 25.5% (n = 123) was observed. Sixty patients who achieved cCR were excluded from final analysis as they underwent organ-preserving surgery (local excision) leaving 63 ypCR patients compared to 359 with RD. On multivariate analysis, combining cCR on MRI and endoscopy with NLR < 5 demonstrated the greatest odds of ypCR on final histological assessment [OR 6.503 (1.594-11.652]) p < 0.001]. This method had the best diagnostic accuracy (AUC = 0.962 95% CI 0.936-0.987), compared to MRI (AUC = 0.711 95% CI 0.650-0.773) or endoscopy (AUC = 0.857 95% CI 0.811-0.902) alone or used together (AUC = 0.926 95% CI 0.892-0.961). CONCLUSION Combining post-NACRT inflammatory markers with restaging MRI and endoscopy findings adds another avenue to aid distinguishing RD from cCR in rectal cancer.
Collapse
|
2
|
Yu X, Liu B, Zhang N, Wang Q, Cheng G. Immune Response: A Missed Opportunity Between Vitamin D and Radiotherapy. Front Cell Dev Biol 2021; 9:646981. [PMID: 33928081 PMCID: PMC8076745 DOI: 10.3389/fcell.2021.646981] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/24/2021] [Indexed: 11/24/2022] Open
Abstract
Radiotherapy (RT) is a mainstay treatment in several types of cancer and acts by mediating various forms of cancer cell death, although it is still a large challenge to enhance therapy efficacy. Radiation resistance represents the main cause of cancer progression, therefore, overcoming treatment resistance is now the greatest challenge for clinicians. Increasing evidence indicates that immune response plays a role in reprogramming the radiation-induced tumor microenvironment (TME). Intriguingly, radiation-induced immunosuppression possibly overwhelms the ability of immune system to ablate tumor cells. This induces an immune equilibrium, which, we hypothesize, is an opportunity for radiosensitizers to make actions. Vitamin D has been reported to act in synergistic with RT by potentiating antiproliferative effect induced by therapeutics. Additionally, vitamin D can also regulate the TME and may even lead to immunostimulation by blocking immunosuppression following radiation. Previous reviews have focused on vitamin D metabolism and epidemiological trials, however, the synergistic effect of vitamin D and existing therapies remains unknown. This review summarizes vitamin D mediated radiosensitization, radiation immunity, and vitamin D-regulated TME, which may contribute to more successful vitamin D-adjuvant radiotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Guanghui Cheng
- Department of Radiation Oncology, China–Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Gennigens C, De Cuypere M, Seidel L, Hermesse J, Barbeaux A, Forget F, Albert A, Jerusalem G, Kridelka F. Correlation between hematological parameters and outcome in patients with locally advanced cervical cancer treated by concomitant chemoradiotherapy. Cancer Med 2020; 9:8432-8443. [PMID: 32954675 PMCID: PMC7666723 DOI: 10.1002/cam4.3465] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 01/28/2023] Open
Abstract
Background Hemoglobin (Hb), white blood cell (WBC), and polymorphonuclear neutrophil (PMN) blood counts may be correlated with outcomes in patients with locally advanced cervical cancer. Methods Hb, WBC, and PMN counts were measured at diagnosis and during concomitant cisplatin‐based chemoradiotherapy (CCRT) in a retrospective sample of 103 patients between 2010 and 2017. Red blood cell (RBC) transfusions were also recorded. The associations between hematological variables and patient overall survival (OS) and recurrence‐free survival (RFS) were assessed by Cox regression models. Results The 3‐year OS and RFS rates were 81.4% and 76.8%, respectively. In addition to tumor size and smoking, OS and RFS were found to be significantly associated with changes in WBC and PMN counts from the first to the last cisplatin cycle. Hb count throughout the treatment and RBC transfusions were not predictive of outcome. Conclusions This study found no association between Hb count or RBC transfusions and outcome. The daily practice of maintaining the Hb count above 12 g/dL during CCRT should be weighed against the potential risks of transfusions. Drops in WBC and PMN counts during treatment positively impacted OS and RFS and could, therefore, serve as biomarkers during CCRT to adapt the follow‐up and consider the need for adjuvant systemic treatments.
Collapse
Affiliation(s)
| | | | - Laurence Seidel
- Department of Biostatistics, CHU Liège and Liège University, Liège, Belgium
| | | | | | - Frédéric Forget
- Department of Medical Oncology, Libramont Hospital, Libramont, Belgium
| | - Adelin Albert
- Department of Biostatistics, CHU Liège and Liège University, Liège, Belgium
| | - Guy Jerusalem
- Department of Medical Oncology, CHU Liège and Liège University, Liège, Belgium
| | - Frédéric Kridelka
- Department of Obstetrics and Gynaecology, CHU Liège and Liège University, Liège, Belgium
| |
Collapse
|
4
|
Yi X, Pei Q, Zhang Y, Zhu H, Wang Z, Chen C, Li Q, Long X, Tan F, Zhou Z, Liu W, Li C, Zhou Y, Song X, Li Y, Liao W, Li X, Sun L, Pei H, Zee C, Chen BT. MRI-Based Radiomics Predicts Tumor Response to Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer. Front Oncol 2019; 9:552. [PMID: 31293979 PMCID: PMC6606732 DOI: 10.3389/fonc.2019.00552] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 06/06/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Conventional methods for predicting treatment response to neoadjuvant chemoradiotherapy (nCRT) in patients with locally advanced rectal cancer (LARC) are limited. Methods: This study retrospectively recruited 134 LARC patients who underwent standard nCRT followed by total mesorectal excision surgery in our institution. Based on pre-operative axial T2-weighted images, machine learning radiomics was performed. A receiver operating characteristic (ROC) curve was performed to test the efficiencies of the predictive model. Results: Among the 134 patients, 32 (23.9%) achieved pathological complete response (pCR), 69 (51.5%) achieved a good response, and 91 (67.9%) achieved down-staging. For prediction of pCR, good-response, and down-staging, the predictive model demonstrated high classification efficiencies, with an AUC value of 0.91 (95% CI: 0.83–0.98), 0.90 (95% CI: 0.83–0.97), and 0.93 (95% CI: 0.87–0.98), respectively. Conclusion: Our machine learning radiomics model showed promise for predicting response to nCRT in patients with LARC. Our predictive model based on the commonly used T2-weighted images on pelvic Magnetic Resonance Imaging (MRI) scans has the potential to be adapted in clinical practice. Novelty and Impact Statements: Methods for predicting the response of the locally advanced rectal cancer (LARC, T3-4, or N+) to neoadjuvant chemoradiotherapy (nCRT) is lacking. In the present study, we developed a new machine learning radiomics method based on T2-weighted images. As a non-invasive tool, this method facilitates prediction performance effectively. It achieves a satisfactory overall diagnostic accuracy for predicting of pCR, good response, and down-staging show an AUC of 0.908, 0.902, and 0.930 in LARC patients, respectively.
Collapse
Affiliation(s)
- Xiaoping Yi
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China.,Postdoctoral Research Workstation of Pathology and Pathophysiology, Basic Medical Sciences, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Pei
- Department of General surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Youming Zhang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Zhu
- Department of Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhongjie Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Chen Chen
- Department of Radiology, Zhuzhou 331 Hospital, Zhuzhou, China
| | - Qingling Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Xueying Long
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Fengbo Tan
- Department of General surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhongyi Zhou
- Department of General surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wenxue Liu
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China
| | - Chenglong Li
- Department of General surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Zhou
- Department of General surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiangping Song
- Department of General surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yuqiang Li
- Department of General surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lunquan Sun
- Postdoctoral Research Workstation of Pathology and Pathophysiology, Basic Medical Sciences, Xiangya Hospital, Central South University, Changsha, China
| | - Haiping Pei
- Department of General surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Chishing Zee
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Bihong T Chen
- Department of Diagnostic Radiology, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
5
|
Taberna M, Oliva M, Mesía R. Cetuximab-Containing Combinations in Locally Advanced and Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma. Front Oncol 2019; 9:383. [PMID: 31165040 PMCID: PMC6536039 DOI: 10.3389/fonc.2019.00383] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/24/2019] [Indexed: 12/29/2022] Open
Abstract
Cetuximab remains to date the only targeted therapy approved for the treatment of head and neck squamous cell carcinoma (HNSCC). The EGFR pathway plays a key role in the tumorigenesis and progression of this disease as well as in the resistance to radiotherapy (RT). While several anti-EGFR agents have been tested in HNSCC, cetuximab, an IgG1 subclass monoclonal antibody against EGFR, is the only drug with proven efficacy for the treatment of both locoregionally-advanced (LA) and recurrent/metastatic (R/M) disease. The addition of cetuximab to radiotherapy is a validated treatment option in LA-HNSCC. However, its use has been limited to patients who are considered unfit for standard of care chemoradiotherapy (CRT) with single agent cisplatin given the lack of direct comparison of these two regimens in randomized phase III trials and the inferiority suggested by metanalysis and phase II studies. The current use of cetuximab in HNSCC is about to change given the recent results from randomized prospective clinical trials in both the LA and R/M setting. Two phase III studies evaluating RT-cetuximab vs. CRT in Human Papillomavirus (HPV)-positive LA oropharyngeal squamous cell carcinoma (De-ESCALaTE and RTOG 1016) showed inferior overall survival and progression-free survival for RT-cetuximab combination, and therefore CRT with cisplatin remains the standard of care in this disease. In the R/M HNSCC, the EXTREME regimen has been the standard of care as first-line treatment for the past 10 years. However, the results from the KEYNOTE-048 study will likely position the anti-PD-1 agent pembrolizumab as the new first line treatment either alone or in combination with chemotherapy in this setting based on PD-L1 status. Interestingly, cetuximab-mediated immunogenicity through antibody dependent cell cytotoxicity (ADCC) has encouraged the evaluation of combined approaches with immune-checkpoint inhibitors in both LA and R/M-HNSCC settings. This article reviews the accumulated evidence on the role of cetuximab in HNSCC in the past decade, offering an overview of its current impact in the treatment of LA and R/M-HNSCC disease and its potential use in the era of immunotherapy.
Collapse
Affiliation(s)
- Miren Taberna
- Medical Oncology Department, Catalan Institute of Oncology, ONCOBELL, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Medicine Department, Barcelona University, Barcelona, Spain
| | - Marc Oliva
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Ricard Mesía
- B-ARGO Group, Medical Oncology Department, Catalan Institute of Oncology, Badalona, Spain
| |
Collapse
|
6
|
Kobiela J, Spychalski P, Marvaso G, Ciardo D, Dell'Acqua V, Kraja F, Błażyńska-Spychalska A, Łachiński AJ, Surgo A, Glynne-Jones R, Jereczek-Fossa BA. Ablative stereotactic radiotherapy for oligometastatic colorectal cancer: Systematic review. Crit Rev Oncol Hematol 2018; 129:91-101. [PMID: 30097241 DOI: 10.1016/j.critrevonc.2018.06.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND SBRT is a novel modality in treatment for oligometastatic colorectal cancer. We aimed to perform a systematic review of results of SBRT in maintaining LC (local control) for CRC liver and lung oligometastases. MATERIALS AND METHODS The review was performed according to PRISMA and PICO guidelines. Database search using keywords: stereotactic, colon, colorectal, cancer, sbrt, sabr returned 457 results. 15 were included in the study. Only cohorts with CRC histology and reported LC were included. RESULTS For liver LC rates ranged from 50% to 100% after 1 year and 32% to 91% after 2 years. BED range 40.5-262.5 Gy (Gray). For lung LC rates ranged from 62% to 92% after 1 one year and from 53% to 92% after 2 years. BED range 51.3-262.5 Gy. CONCLUSIONS SBRT of oligometastatic CRC offers high LC with low morbidity and toxicity. It requires more observational studies and randomized trials but available data on clinical efficacy is promising, however not yet matured.
Collapse
Affiliation(s)
- J Kobiela
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, Gdansk, Poland
| | - P Spychalski
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, Gdansk, Poland.
| | - G Marvaso
- Department of Radiotherapy, European Institute of Oncology, Milan, Italy
| | - D Ciardo
- Department of Radiotherapy, European Institute of Oncology, Milan, Italy
| | - V Dell'Acqua
- Department of Radiotherapy, European Institute of Oncology, Milan, Italy
| | - F Kraja
- Department of Oncology, University Hospital Centre "Mother Theresa", Tirana, Albania
| | - A Błażyńska-Spychalska
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, Gdansk, Poland
| | - A J Łachiński
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, Gdansk, Poland
| | - A Surgo
- Department of Radiotherapy, European Institute of Oncology, Milan, Italy
| | - R Glynne-Jones
- Mount Vernon Centre for Cancer Treatment, Northwood, Middlesex, HA6 2RN, UK
| | - B A Jereczek-Fossa
- Department of Radiotherapy, European Institute of Oncology, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
7
|
Barbetta A, Nobel TB, Sihag S, Hsu M, Tan KS, Bains MS, Isbell JM, Janjigian YY, Wu AJ, Bott MJ, Jones DR, Molena D. Neutrophil to Lymphocyte Ratio as Predictor of Treatment Response in Esophageal Squamous Cell Cancer. Ann Thorac Surg 2018; 106:864-871. [PMID: 29738752 DOI: 10.1016/j.athoracsur.2018.04.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 03/07/2018] [Accepted: 04/02/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND The aim of this study was to assess the difference (Δ) in neutrophil to lymphocyte ratio (NLR), before and after chemoradiotherapy, as a predictor of treatment response and a prognostic factor for recurrence and disease-free survival in patients with esophageal squamous cell cancer treated with chemoradiotherapy with or without surgery. METHODS Patients with locally advanced esophageal squamous cell cancer treated with chemoradiation with and without surgery who had a complete blood count before and after chemoradiotherapy were included. Pretreatment and posttreatment NLR were calculated by dividing the absolute neutrophil count by the absolute lymphocyte count. The ΔNLR was defined as posttreatment minus pretreatment NLR. Characteristics were evaluated for association with ΔNLR using the Wilcoxon signed rank test or the Kruskal-Wallis test. Risk of recurrence and disease-free survival were evaluated using Gray's and the log rank tests, respectively. RESULTS We included 217 patients. Of them, 133 patients (61.3%) received only chemoradiotherapy and 84 (38.7%) underwent surgery after chemoradiotherapy. Among the surgical patients, 43% with pathologic complete response showed significantly lower median ΔNLR than patients with residual disease (-0.03 versus 1.04, p = 0.004). High ΔNLR was a negative predictor of treatment response (odds ratio 0.77, 95% confidence interval: 0.62 to 0.9, p = 0.004). A significant association between high ΔNLR and increased risk of recurrence was also identified. CONCLUSIONS The ΔNLR was inversely related to pathologic complete response and associated with risk of recurrence. This simple test, in concert with other clinical tools, can help identify patients with pathologic complete response.
Collapse
Affiliation(s)
- Arianna Barbetta
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tamar B Nobel
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Smita Sihag
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Meier Hsu
- Biostatistics Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kay See Tan
- Biostatistics Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Manjit S Bains
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James M Isbell
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yelena Y Janjigian
- Gastrointestinal Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Abraham J Wu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew J Bott
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Molena
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
8
|
Noh OK, Oh SY, Kim YB, Suh KW. Prognostic Significance of Lymphocyte Counts in Colon Cancer Patients Treated with FOLFOX Chemotherapy. World J Surg 2018; 41:2898-2905. [PMID: 28707088 DOI: 10.1007/s00268-017-4104-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE There is increasing interest in immune function in combination with chemotherapy for cancer treatment. However, the effects of chemotherapy on the human immune system remain to be determined. The aim of this study was to investigate the prognostic impact of lymphocyte and neutrophil counts in colon cancer patients who were treated with curative surgery and adjuvant chemotherapy. METHODS Two hundred thirty-one patients with colon cancers who underwent curative surgery and FOLFOX adjuvant chemotherapy between November 2005 and December 2011 were included. Oncologic outcomes were analyzed with neutrophil count, lymphocyte count, and neutrophil-to-lymphocyte ratio (NLR) before and after chemotherapy. RESULTS The 5-year DFS rate was lower in colon cancer patients with low lymphocyte count during chemotherapy (61.9 vs. 76.7%, P = 0.026). Cox multivariate analysis demonstrated that low lymphocyte count during chemotherapy was independently associated with poor disease-free survival (HR 1.829; 95% CI 1.096-3.050; P = 0.021) in colon cancer patients who underwent FOLFOX adjuvant chemotherapy. CONCLUSION Lymphocyte count during chemotherapy is a strong predictor of worse disease-free survival in colon cancer patients who have undergone FOLFOX adjuvant chemotherapy.
Collapse
Affiliation(s)
- O Kyu Noh
- Department of Radiation Oncology, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Korea
| | - Seung Yeop Oh
- Department of Surgery, Ajou University School of Medicine, San 5, Woncheon-dong, Yeongtong-gu, Suwon, 443-721, Korea.
| | - Young Bae Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Kwang Wook Suh
- Department of Surgery, Ajou University School of Medicine, San 5, Woncheon-dong, Yeongtong-gu, Suwon, 443-721, Korea
| |
Collapse
|
9
|
Vandenabeele P, Vandecasteele K, Bachert C, Krysko O, Krysko DV. Immunogenic Apoptotic Cell Death and Anticancer Immunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 930:133-49. [PMID: 27558820 DOI: 10.1007/978-3-319-39406-0_6] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
For many years it has been thought that apoptotic cells rapidly cleared by phagocytic cells do not trigger an immune response but rather have anti-inflammatory properties. However, accumulating experimental data indicate that certain anticancer therapies can induce an immunogenic form of apoptosis associated with the emission of damage-associated molecular patterns (DAMPs), which function as adjuvants to activate host antitumor immune responses. In this review, we will first discuss recent advances and the significance of danger signaling pathways involved in the emission of DAMPs, including calreticulin, ATP, and HMGB1. We will also emphasize that switching on a particular signaling pathway depends on the immunogenic cell death stimulus. Further, we address the role of ER stress in danger signaling and the classification of immunogenic cell death inducers in relation to how ER stress is triggered. In the final part, we discuss the role of radiotherapy-induced immunogenic apoptosis and the relationship of its immunogenicity to the fraction dose and concomitant chemotherapy.
Collapse
Affiliation(s)
- Peter Vandenabeele
- Molecular Signalling and Cell Death Unit, Inflammation Research Center, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Methusalem program, Ghent University, Ghent, Belgium
| | - Katrien Vandecasteele
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Claus Bachert
- The Upper Airway Research Laboratory, Department of Oto-Rhino-Laryngology, Ghent University Hospital, Ghent, Belgium
| | - Olga Krysko
- The Upper Airway Research Laboratory, Department of Oto-Rhino-Laryngology, Ghent University Hospital, Ghent, Belgium
| | - Dmitri V Krysko
- Molecular Signalling and Cell Death Unit, Inflammation Research Center, VIB, Ghent, Belgium. .,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
10
|
Liu X, Li M, Zhao F, Zhu Y, Luo Y, Kong L, Zhu H, Zhang Y, Shi F, Yu J. The lymphocyte-monocyte ratio predicts tumor response and survival in patients with locally advanced esophageal cancer who received definitive chemoradiotherapy. Onco Targets Ther 2017; 10:871-877. [PMID: 28243122 PMCID: PMC5317323 DOI: 10.2147/ott.s124915] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Background The lymphocyte–monocyte ratio (LMR), a simple biomarker that can reflect the antitumor immune response of the host, has been associated with patient prognosis in several solid tumors. The aim of this study was to evaluate whether LMR can predict clinical tumor response and prognosis in patients with locally advanced esophageal squamous cell carcinoma (ESCC) who received definitive chemoradiotherapy (CRT). Patients and methods A total of 162 advanced ESCC patients treated at our institution between January 2012 and December 2013 were retrospectively recruited for analysis. Patients were treated with a platinum-based bimodal cytotoxic drug chemotherapy and concurrent radiation therapy. The LMR was calculated from blood counts in samples collected prior to treatment initiation. The predictive value of LMR for clinical tumor response and prognosis was examined. Results The LMR before CRT was significantly higher in 48 patients who achieved clinical complete response (CR) compared to that in patients who did not achieve clinical CR (4.89±1.17 vs 3.87±1.29, P<0.001). Compared to their matched counterparts, patients in the high LMR group (LMR >4.02) showed a good clinical tumor response (P<0.05). A significant independent association between a high pretreatment LMR and better outcomes was identified in a multivariate analysis for progression-free survival (PFS; hazard ratio [HR]=2.17; P<0.001) and overall survival (OS; HR=2.02; P=0.002). Conclusion In ESCC patients, a high LMR before treatment, which indicates a robust host immune system, is associated with both a good clinical tumor response after definitive CRT and favorable prognosis.
Collapse
Affiliation(s)
- Xuemei Liu
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Minghuan Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Fen Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Yingming Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Yijun Luo
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Li Kong
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Hui Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Yan Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Fang Shi
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| |
Collapse
|
11
|
Formenti SC, Golden EB, Goldberg JD, Li X, Taff J, Fenton-Kerimian MB, Chandrasekhar S, Demaria S, Novik Y. Results of a phase I-II study of adjuvant concurrent carboplatin and accelerated radiotherapy for triple negative breast cancer. Oncoimmunology 2016; 6:e1274479. [PMID: 28405497 DOI: 10.1080/2162402x.2016.1274479] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/12/2016] [Accepted: 12/14/2016] [Indexed: 12/31/2022] Open
Abstract
Purpose: To determine feasibility and explore the clinical efficacy of concurrent radiotherapy and carboplatin as adjuvant treatment of triple negative breast cancer (TNBC). Patients and Methods: Women with Stage I-II TNBC were treated after surgery in a phase I-II prospective trial [NCT01289353]. Weekly carboplatin (AUC = 2.0) was delivered for 6 weeks. Concurrent radiotherapy was delivered in the prone position during weeks 2-4, for a total dose of 40.5 Gy in 15 fractions to the breast, and 46.5 Gy in 17 fractions to the tumor bed. Adverse events (AE) were assessed weekly during treatment, once at 45-60 d, and every 6 mo thereafter, using the Common Terminology Criteria for AE (CTCAE) v3.0. Results: A total of 39 patients accrued and 36 received treatment. Eight patients (22%, exact 95% CI: 10%, 39%) developed grade 2 or greater acute radiation dermatitis. Overall, grade 2 AE were seen in nine and grade 3 in two patients. Twenty-three patients (64%) received additional adjuvant chemotherapy. With a median follow-up of 48 mo, 34/36 (94%) are alive and disease free. One patient died of pulmonary failure with possible but unproven breast cancer recurrence, and one patient died of pelvic malignancy. One patient recurred locally and is alive and disease free after surgical management. Brisk lymphocytic infiltrate was present pre-treatment in 39% of 18 patients with evaluable tumor. Conclusions: Adjuvant concurrent carboplatin and prone accelerated radiotherapy is a well-tolerated and promising treatment of early stage TNBC. The observed 3% compares favorably with the expected 30% recurrence rate within 1-4 y from treatment, warranting further studies.
Collapse
Affiliation(s)
- Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medicine , New York, NY, USA
| | - Encouse B Golden
- Department of Radiation Oncology, Weill Cornell Medicine , New York, NY, USA
| | - Judith D Goldberg
- Departments of Population Health and Environmental Medicine, New York University School of Medicine , New York, NY, USA
| | - Xiaochun Li
- Departments of Population Health and Environmental Medicine, New York University School of Medicine , New York, NY, USA
| | - Jessica Taff
- Department of Medicine, New York University School of Medicine , New York, NY, USA
| | | | | | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine , New York, NY, USA
| | - Yelena Novik
- Department of Medicine, New York University School of Medicine , New York, NY, USA
| |
Collapse
|
12
|
Sohun M, Shen H. The implication and potential applications of high-mobility group box 1 protein in breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:217. [PMID: 27386491 PMCID: PMC4916368 DOI: 10.21037/atm.2016.05.36] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/14/2016] [Indexed: 01/09/2023]
Abstract
High-mobility group box 1 protein (HMGB1) is a highly conserved, non-histone and ubiquitous chromosomal protein found enriched in active chromatin forming part of the high mobility group family of proteins and is encoded by the HMGB1 gene (13q12) in human beings. It has various intranuclear and extracellular functions. It plays an important role in the pathogenesis of many diseases including cancer. In 2012, there was approximately 1.67 million new breast cancer cases diagnosed which makes it the second most frequent cancer in the world after lung cancer (25% of all cancers) and the commonest cancer among women. Both pre-clinical and clinical studies have suggested that HMGB1 might be a useful target in the management of breast cancer. This review summarises the structure and functions of HMGB1 and its dual role in carcinogenesis both as a pro-tumorigenic and anti-tumorigenic factor. It also sums up evidence from in vitro and in vivo studies using breast cancer cell lines and samples which demonstrate its influence in radiotherapy, chemotherapy and hormonal therapy in breast cancer. It may have particular importance in HER2 positive and metastatic breast cancer. It might pave the way for new breast cancer treatments through development of novel drugs, use of microRNAs (miRNAs), targeting breast cancer stem cells (CSCs) and breast cancer immunotherapy. It may also play a role in determining breast cancer prognosis. Thus HMGB1 may open up novel avenues in breast cancer management.
Collapse
Affiliation(s)
- Moonindranath Sohun
- Department of Oncology, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212013, China
| | - Huiling Shen
- Department of Oncology, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
13
|
Kumari A, Garnett-Benson C. Effector function of CTLs is increased by irradiated colorectal tumor cells that modulate OX-40L and 4-1BBL and is reversed following dual blockade. BMC Res Notes 2016; 9:92. [PMID: 26872462 PMCID: PMC4752774 DOI: 10.1186/s13104-016-1914-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/03/2016] [Indexed: 01/10/2023] Open
Abstract
Background Sub-lethal doses of ionizing radiation (IR) can alter the phenotype of target tissue by modulating genes that influence effector T cell activity. Previous studies indicate that cancer cells respond to radiation by up-regulating surface expression of death receptors, cell adhesion molecules and tumor-associated antigens (TAA). However, there is limited information available regarding how T cells themselves are altered following these interactions with irradiated tumor cells. Methods Here, several human colorectal tumor cell lines were exposed to radiation (0–10 Gy) in vitro and changes in the expression of molecules costimulatory to effector T cells (4-1BBL, OX-40L, CD70, ICOSL) were examined by flow cytometry. T cell effector function was assessed to determine if changes in these proteins were directly related to the changes in T cell function. Results We found OX-40L and 4-1BBL to be the most consistently upregulated proteins on the surface of colorectal tumor cells post-IR while ICOSL and CD70 remained largely unaltered. Expression of these gene products correlated with enhanced killing of irradiated human colorectal tumor cells by TAA-specific T-cells. Importantly, blocking of both OX-40L and 4-1BBL reversed radiation-enhanced T-cell killing of human tumor targets as well as T-cell survival and activation. Conclusions Overall, results of this study suggest that, beyond simply rendering tumor cells more sensitive to immune attack, radiation can be used to specifically modulate expression of genes that directly stimulate effector T cell activity.
Collapse
Affiliation(s)
- Anita Kumari
- Department of Biology, Georgia State University, 161 Jesse Hill Jr. Dr, Atlanta, GA, 30303, USA.
| | - Charlie Garnett-Benson
- Department of Biology, Georgia State University, 161 Jesse Hill Jr. Dr, Atlanta, GA, 30303, USA.
| |
Collapse
|
14
|
Abstract
Advances in understanding the mechanisms that underlie the interplay between radiation-invoked immune responses and tumor regression are underway. Emerging applications of local radiotherapy as an immunologic adjuvant have provided radiation oncologists with a method for converting malignant cells into endogenous anticancer vaccines. The dispersion of radiotherapy-induced immune-stimulating tumor antigens released from dying tumor cells into the surrounding milieu (known as immunogenic cell death, Fig. 1), is one such exploitable process that contributes to the propagation of antitumor immunity. Downstream components of the immune system may suppress, promote, or ambiguously affect antitumoral responses. Additionally, host, tumor, and treatment-related characteristics govern the significance of these signals, thereby dictating therapeutic outcomes. Herein, we review the process of radiotherapy-induced immunogenic cell death and its role in generating an in situ vaccine to help refine radioimmunotherapy-based protocols.
Collapse
Affiliation(s)
- Encouse B Golden
- Department of Radiation Oncology, New York University School of Medicine, New York, NY.
| | - Lionel Apetoh
- INSERM U866, Dijon, France; Université de Bourgogne, Dijon, France; Centre Georges François Leclerc, Dijon, France
| |
Collapse
|
15
|
Kao CJ, Wurz GT, Lin YC, Vang DP, Griffey SM, Wolf M, DeGregorio MW. Assessing the Effects of Concurrent versus Sequential Cisplatin/Radiotherapy on Immune Status in Lung Tumor-Bearing C57BL/6 Mice. Cancer Immunol Res 2015; 3:741-50. [PMID: 25672395 DOI: 10.1158/2326-6066.cir-14-0234] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/04/2015] [Indexed: 11/16/2022]
Abstract
Concurrent and sequential cisplatin-based chemoradiotherapy regimens are standard therapeutic approaches in cancer treatment. Recent clinical data suggest that these different dosing schedules may adversely affect antigen-specific immunotherapy. The goal of the present preclinical study was to explore the effects of concurrent and sequential cisplatin/radiotherapy on immune status in a lung cancer mouse model. A total of 150 C57BL/6 mice were randomized into six treatment groups: control; 8 Gy thoracic radiotherapy (dose schedules 1 and 2); cisplatin 2.5 mg/kg i.p.; cisplatin + radiotherapy (concurrent); and cisplatin + radiotherapy (sequential; n = 25, all groups). At the end of the study (week 41), serum cytokines were assessed by multiplex immunoassay, surface markers of spleen-derived lymphocytes were assessed by immunostaining and flow cytometry, lung tumor expression of programmed death ligands 1 and 2 (PD-L1/2) was evaluated by immunohistochemistry, and miRNA profiling was performed in serum and lymphocytes by quantitative real-time PCR. Lung whole mounts were prepared to assess treatment effects on lung tumor foci formation. The results showed that sequential chemoradiotherapy (two cycles of cisplatin followed by 8 Gy radiotherapy) had equivalent antitumor activity as concurrent therapy. However, sequential cisplatin/radiotherapy resulted in significant differences in several immune response biomarkers, including regulatory T cells, miR-29c, expression of costimulatory molecule CD28, and serum IFNγ. PD-L1 and PD-L2 were strongly expressed in tumor foci, but no trend was seen between groups. These results suggest that monitoring immune status may be necessary when designing treatment regimens combining immunotherapy with chemoradiotherapy.
Collapse
Affiliation(s)
- Chiao-Jung Kao
- Division of Hematology and Oncology, Department of Internal Medicine, University of California, Davis, Sacramento, California
| | - Gregory T Wurz
- Division of Hematology and Oncology, Department of Internal Medicine, University of California, Davis, Sacramento, California
| | - Yi-Chen Lin
- Division of Hematology and Oncology, Department of Internal Medicine, University of California, Davis, Sacramento, California
| | - Daniel P Vang
- Division of Hematology and Oncology, Department of Internal Medicine, University of California, Davis, Sacramento, California
| | - Stephen M Griffey
- Comparative Pathology Laboratory, UC Davis School of Veterinary Medicine, University of California, Davis, Davis, California
| | - Michael Wolf
- ImmunoOncology, Merck Serono Research, Merck KGaA, Darmstadt, Germany
| | - Michael W DeGregorio
- Division of Hematology and Oncology, Department of Internal Medicine, University of California, Davis, Sacramento, California.
| |
Collapse
|
16
|
Kim IY, You SH, Kim YW. Neutrophil-lymphocyte ratio predicts pathologic tumor response and survival after preoperative chemoradiation for rectal cancer. BMC Surg 2014; 14:94. [PMID: 25406793 PMCID: PMC4242604 DOI: 10.1186/1471-2482-14-94] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 08/01/2014] [Indexed: 02/08/2023] Open
Abstract
Background Neutrophil-lymphocyte ratio (NLR) reflects the balance between pro- and anti-tumor immune activities. We evaluated whether NLR is associated with pathologic tumor response and prognosis in rectal cancer patients that underwent preoperative chemoradiaton therapy (CRT) with surgery. Methods One hundred two patients with rectal cancer that were treated by preoperative CRT followed by surgery were enrolled. A total of 50.4 GY of radiation and 5-FU-based chemotherapy were delivered. An NLR ≥ 3 was considered to be elevated. Pathologic tumor response based on ypTNM stage was categorized into two groups, good response (n = 35, pathologic complete response and ypTNM I) and poor response groups (n = 67, ypTNM II, III, and IV). Results Twenty-five patients (24.5%) had elevated NLR. Multivariate analysis showed that an elevated CEA level (p = 0.001), larger tumor (p = 0.03), and elevated NLR (p = 0.04) were significant predictors for a poor response. Poor pathological tumor response and elevated NLR were risk factors for cancer-specific and recurrence-free survivals. Conclusion An elevated NLR before CRT can be used as predictors for poor tumor response and unfavorable prognostic factors. Dominant pro-tumor activities of neutrophils or reduced anti-tumor immune response by lymphocytes, as determined by NLR, may have a impact on poor tumor response and unfavorable prognosis. Electronic supplementary material The online version of this article (doi:10.1186/1471-2482-14-94) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Young Wan Kim
- Department of Surgery, Yonsei University Wonju College of Medicine, 162 Ilsan-dong, Wonju-si, Gangwon-do (220-701), Korea.
| |
Collapse
|
17
|
Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology 2014; 3:e28518. [PMID: 25071979 PMCID: PMC4106151 DOI: 10.4161/onci.28518] [Citation(s) in RCA: 443] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 03/10/2014] [Accepted: 03/12/2014] [Indexed: 12/13/2022] Open
Abstract
Established tumors are typified by an immunosuppresive microenvironment. Countering this naturally occurring phenomenon, emerging evidence suggests that radiation promotes a proimmunogenic milieu within the tumor capable of stimulating host cancer-specific immune responses. Three cryptic immunogenic components of cytotoxic-agent induced cell death—namely, calreticulin cell surface exposure, the release of high mobility group box 1 (HMGB1) protein, and the liberation of ATP—have been previously shown to be critical for dendritic cell (DC) activation and effector T-cell priming. Thus, these immune-mobilizing components commonly presage tumor rejection in response to treatment. We initially set out to address the hypothesis that radiation-induced immunogenic cell death (ICD) is dose-dependent. Next, we hypothesized that radiation would enhance chemotherapy-induced ICD when given concomitantly, as suggested by the favorable clinical outcomes observed in response to analogous concurrent chemoradiation regimens. Thus, we designed an in vitro assay to examine the 3 hallmark features of ICD at clinically relevant doses of radiation. We then tested the immunogenic-death inducing effects of radiation combined with carboplatin or paclitaxel, focusing on these combinations to mimic chemoradiation regimens actually used in clinical trials of early stage triple negative [NCT0128953/NYU-10–01969] and locally advanced [NYU-06209] breast cancer patients, respectively. Despite the obvious limitations of an in vitro model, radiotherapy produced both a dose-dependent induction and chemotherapeutic enhancement of ICD. These findings provide preliminary evidence that ICD stimulated by either high-dose radiotherapy alone, or concurrent chemoradiation regimens, may contribute to the establishment of a peritumoral proimmunogenic milieu.
Collapse
Affiliation(s)
- Encouse B Golden
- Department of Radiation Oncology; New York University School of Medicine; New York, NY USA
| | - Derek Frances
- Department of Radiation Oncology; New York University School of Medicine; New York, NY USA
| | - Ilenia Pellicciotta
- Department of Radiation Oncology; New York University School of Medicine; New York, NY USA
| | - Sandra Demaria
- Department of Radiation Oncology; New York University School of Medicine; New York, NY USA ; Department of Pathology; New York University School of Medicine; New York, NY USA
| | | | - Silvia C Formenti
- Department of Radiation Oncology; New York University School of Medicine; New York, NY USA
| |
Collapse
|
18
|
Ladoire S, Hannani D, Vetizou M, Locher C, Aymeric L, Apetoh L, Kepp O, Kroemer G, Ghiringhelli F, Zitvogel L. Cell-death-associated molecular patterns as determinants of cancer immunogenicity. Antioxid Redox Signal 2014; 20:1098-116. [PMID: 23394620 DOI: 10.1089/ars.2012.5133] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Accumulating evidence indicates that the success of some anticancer treatments (select chemotherapies or radiotherapy or trastuzumab) could be related to the stimulation of an anticancer immune response through the induction of an immunogenic tumor cell death (ICD). RECENT ADVANCES Preclinical data revealed that dying tumor cells can emit a series of danger signals (so-called "cell-death-associated molecular patterns" (CDAMP)) that will dictate the recruitment and activation of specific inflammatory phagocytes. Hence, tumor cells succumbing to ICD are characterized by specific metabolic and molecular changes that will trigger a hierarchy of polarizing cytokine-producing cells, culminating in the recruitment and reactivation of antitumor interferon-γ-producing effector T cells which contribute to the success of cytotoxic treatments. CRITICAL ISSUES In this review, we summarize the molecular and cellular bases of this ICD, underscoring the crucial role of high mobility group box 1 protein (HMGB1) and adenosine tri-phosphate, both of which are released from dying tumor cells during ICD and are implicated in the chemotherapy-elicited anticancer immune response. FUTURE DIRECTIONS We discuss here how such CDAMP could serve as predictive biomarkers that could discriminate immunogenic from nonimmunogenic anti-cancer compounds, and, in case of deficiency, could be compensated by surrogate products to ameliorate the success rate of conventional anticancer treatment modalities.
Collapse
Affiliation(s)
- Sylvain Ladoire
- 1 Institut National de la Santé et de la Recherche Médicale , Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Lung cancer has traditionally been considered relatively resistant to immunotherapies. However, recent advances in the understanding of tumor-associated antigens, anti-tumor immune responses, and tumor immunosuppression mechanisms have resulted in a number of promising immunomodulatory therapies such as vaccines and checkpoint inhibitors. Locally advanced non-small cell lung cancer is an optimal setting for these treatments because standard therapies such as surgery, radiation, and chemotherapy may enhance anti-tumor immune effects by debulking the tumor, increasing tumor antigen presentation, and promoting T-cell response and trafficking. Clinical trials incorporating immunomodulatory agents into combined modality therapy of locally advanced non-small cell lung cancer have shown promising results. Future challenges include identifying biomarkers to predict those patients most likely to benefit from this approach, radiographic assessment of treatment effects, the timing and dosing of combined modality treatment including immunotherapies, and avoidance of potentially overlapping toxicities.
Collapse
|
20
|
Formenti SC, Demaria S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. J Natl Cancer Inst 2013; 105:256-65. [PMID: 23291374 DOI: 10.1093/jnci/djs629] [Citation(s) in RCA: 787] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The therapeutic application of ionizing radiation has been largely based on its cytocidal power combined with the ability to selectively target tumors. Radiotherapy effects on survival of cancer patients are generally interpreted as the consequence of improved local control of the tumor, directly decreasing systemic spread. Experimental data from multiple cancer models have provided sufficient evidence to propose a paradigm shift, whereby some of the effects of ionizing radiation are recognized as contributing to systemic antitumor immunity. Recent examples of objective responses achieved by adding radiotherapy to immunotherapy in metastatic cancer patients support this view. Therefore, the traditional palliative role of radiotherapy in metastatic disease is evolving into that of a powerful adjuvant for immunotherapy. This combination strategy adds to the current anticancer arsenal and offers opportunities to harness the immune system to extend survival, even among metastatic and heavily pretreated cancer patients. We briefly summarize key evidence supporting the role of radiotherapy as an immune adjuvant. A critical appraisal of the current status of knowledge must include potential immunosuppressive effects of radiation that can hamper its capacity to convert the irradiated tumor into an in situ, individualized vaccine. Moreover, we discuss some of the current challenges to translate this knowledge to the clinic as more trials testing radiation with different immunotherapies are proposed.
Collapse
Affiliation(s)
- Silvia C Formenti
- Department of Radiation Oncology, New York University School of Medicine, 160 E 34th St, New York, NY 10016, USA.
| | | |
Collapse
|
21
|
Mason KA, Hunter NR. CpG plus radiotherapy: a review of preclinical works leading to clinical trial. Front Oncol 2012; 2:101. [PMID: 22912936 PMCID: PMC3418655 DOI: 10.3389/fonc.2012.00101] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 07/28/2012] [Indexed: 02/05/2023] Open
Abstract
Studies performed three decades ago in our laboratory supported the hypothesis that radiation efficacy may be augmented by bacterial extracts that stimulate non-specific systemic antitumor immune responses. Application to the clinic was halted by unacceptable side effects and toxicities resulting from exposure to whole bacterial pathogens. Later scientific advances demonstrated that DNA isolated from bacteria was immunostimulatory and could be reproduced with synthetic oligodeoxynucleotides (ODNs), thus fueling the transition from bugs to drugs. Unmethylated CpG motifs within bacterial DNA induce activation of Toll-like receptor 9 and subsequently activate antigen-specific cellular immune responses. CpG ODNs have demonstrated favorable toxicity profiles in phase I clinical trials. We showed that this potent immunoadjuvant can be used in combination with radiation therapy to enhance local and systemic responses of several murine tumors. Studies demonstrated that enhanced tumor response is mediated in part by the host immune system. Antitumor efficacy was diminished in immunocompromised mice. Animals cured by combination of radiation and CpG ODN were resistant to subsequent tumor rechallenge. This body of work contributes to our understanding of the dynamic interplay between tumor irradiation and the host immune system and may facilitate translation to clinical trials.
Collapse
Affiliation(s)
- Kathy A Mason
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center Houston, TX, USA
| | | |
Collapse
|
22
|
Golden EB, Pellicciotta I, Demaria S, Barcellos-Hoff MH, Formenti SC. The convergence of radiation and immunogenic cell death signaling pathways. Front Oncol 2012; 2:88. [PMID: 22891162 PMCID: PMC3413017 DOI: 10.3389/fonc.2012.00088] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 07/18/2012] [Indexed: 11/13/2022] Open
Abstract
Ionizing radiation (IR) triggers programmed cell death in tumor cells through a variety of highly regulated processes. Radiation-induced tumor cell death has been studied extensively in vitro and is widely attributed to multiple distinct mechanisms, including apoptosis, necrosis, mitotic catastrophe (MC), autophagy, and senescence, which may occur concurrently. When considering tumor cell death in the context of an organism, an emerging body of evidence suggests there is a reciprocal relationship in which radiation stimulates the immune system, which in turn contributes to tumor cell kill. As a result, traditional measurements of radiation-induced tumor cell death, in vitro, fail to represent the extent of clinically observed responses, including reductions in loco-regional failure rates and improvements in metastases free and overall survival. Hence, understanding the immunological responses to the type of radiation-induced cell death is critical. In this review, the mechanisms of radiation-induced tumor cell death are described, with particular focus on immunogenic cell death (ICD). Strategies combining radiotherapy with specific chemotherapies or immunotherapies capable of inducing a repertoire of cancer specific immunogens might potentiate tumor control not only by enhancing cell kill but also through the induction of a successful anti-tumor vaccination that improves patient survival.
Collapse
Affiliation(s)
- Encouse B Golden
- Department of Radiation Oncology, New York University New York, NY, USA
| | | | | | | | | |
Collapse
|
23
|
Is the combination of Cetuximab with chemo-radiotherapy regimens worthwhile in the treatment of locally advanced head and neck cancer? A review of current evidence. Crit Rev Oncol Hematol 2012; 85:112-20. [PMID: 22743346 DOI: 10.1016/j.critrevonc.2012.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Revised: 05/12/2012] [Accepted: 05/31/2012] [Indexed: 12/27/2022] Open
Abstract
The administration of Cetuximab in combination with radiotherapy and chemotherapy has shown clear survival improvements within the locally advanced and the relapsed/metastatic settings respectively. These results have provided the clinical rational for the inclusion of Cetuximab into chemo-radiation regimens. Trials assessing the combination of Cetuximab with induction chemotherapy, concomitant chemo-radiotherapy or both are reviewed. Taken together, their results suggest that the addition of Cetuximab is promising in trials of induction chemotherapy, showing almost uniformly response rates higher than historical controls. In combination with concomitant hyperfractionated radiotherapy and Cisplatin the results of the RTOG 0522 trial do not suggest any benefit. However a positive effect cannot be excluded with other schedules. Although feasibility has been universally suggested, adding Cetuximab implies some toxicity enhancement. Single local and systemic toxicities are more frequent and supposedly the overall treatment intensity is increased. Moreover the drug-specific toxicities are potentially severe and deserve timely recognition and management.
Collapse
|
24
|
Haynes NM, Smyth MJ. Immunotherapeutic strategies as adjuncts to local radiotherapy. Immunotherapy 2012; 4:129-31. [PMID: 22339453 DOI: 10.2217/imt.11.155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
25
|
Ifeadi V, Garnett-Benson C. Sub-lethal irradiation of human colorectal tumor cells imparts enhanced and sustained susceptibility to multiple death receptor signaling pathways. PLoS One 2012; 7:e31762. [PMID: 22389673 PMCID: PMC3289623 DOI: 10.1371/journal.pone.0031762] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 01/18/2012] [Indexed: 01/01/2023] Open
Abstract
Background Death receptors (DR) of the TNF family function as anti-tumor immune effector molecules. Tumor cells, however, often exhibit DR-signaling resistance. Previous studies indicate that radiation can modify gene expression within tumor cells and increase tumor cell sensitivity to immune attack. The aim of this study is to investigate the synergistic effect of sub-lethal doses of ionizing radiation in sensitizing colorectal carcinoma cells to death receptor-mediated apoptosis. Methodology/Principal Findings The ability of radiation to modulate the expression of multiple death receptors (Fas/CD95, TRAILR1/DR4, TRAILR2/DR5, TNF-R1 and LTβR) was examined in colorectal tumor cells. The functional significance of sub-lethal doses of radiation in enhancing tumor cell susceptibility to DR-induced apoptosis was determined by in vitro functional sensitivity assays. The longevity of these changes and the underlying molecular mechanism of irradiation in sensitizing diverse colorectal carcinoma cells to death receptor-mediated apoptosis were also examined. We found that radiation increased surface expression of Fas, DR4 and DR5 but not LTβR or TNF-R1 in these cells. Increased expression of DRs was observed 2 days post-irradiation and remained elevated 7-days post irradiation. Sub-lethal tumor cell irradiation alone exhibited minimal cell death, but effectively sensitized three of three colorectal carcinoma cells to both TRAIL and Fas-induced apoptosis, but not LTβR-induced death. Furthermore, radiation-enhanced Fas and TRAIL-induced cell death lasted as long as 5-days post-irradiation. Specific analysis of intracellular sensitizers to apoptosis indicated that while radiation did reduce Bcl-XL and c-FLIP protein expression, this reduction did not correlate with the radiation-enhanced sensitivity to Fas and/or TRAIL mediated apoptosis among the three cell types. Conclusions/Significance Irradiation of tumor cells can overcome Fas and TRAIL resistance that is long lasting. Overall, results of these investigations suggest that non-lethal doses of radiation can be used to make human tumors more amenable to attack by anti-tumor effector molecules and cells.
Collapse
Affiliation(s)
| | - Charlie Garnett-Benson
- Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
26
|
Vicente Conesa MA, Garcia-Martinez E, Gonzalez Billalabeitia E, Chaves Benito A, Garcia Garcia T, Vicente Garcia V, Ayala de la Peña F. Predictive value of peripheral blood lymphocyte count in breast cancer patients treated with primary chemotherapy. Breast 2011; 21:468-74. [PMID: 22119767 DOI: 10.1016/j.breast.2011.11.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 09/21/2011] [Accepted: 11/03/2011] [Indexed: 01/30/2023] Open
Abstract
Peripheral blood lymphocyte (PBL) count may reflect the immune status of cancer patients. We retrospectively analyzed the predictive and prognostic impact of baseline and post-chemotherapy PBL counts in a homogeneous group of 103 breast cancer patients treated with neoadjuvant chemotherapy (anthracyclines and taxanes). In univariate analysis, baseline PBL under 1500 × 10(6)/L (p = 0.013; hazard ratio [HR]: 2.80, 95%CI 1.24-6.61), and PBL decrease >200 × 10(6)/L after the first cycle of chemotherapy (p = 0.047; HR: 2.82, 95%CI 1.01-7.86) were significantly related to disease free survival. In multivariate analysis, both baseline PBL count less than 1500 × 10(6)/L (p = 0.034; HR: 3.32, 95%CI 1.09-10.02) and PBL decrease >200 × 10(6)/L after first cycle (p = 0.032; HR: 3.25, 95%CI 1.10-9.56) showed independent prognostic value for worse disease free survival. No effect was observed for overall survival. Our data support the relevance of pre- and post-chemotherapy PBL for breast cancer recurrence after neoadjuvant chemotherapy.
Collapse
|
27
|
Preoperative concurrent paclitaxel-radiation in locally advanced breast cancer: pathologic response correlates with five-year overall survival. Breast Cancer Res Treat 2010; 124:723-32. [PMID: 20878462 DOI: 10.1007/s10549-010-1181-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 09/14/2010] [Indexed: 12/31/2022]
Abstract
We have previously demonstrated high pathologic response rates after neoadjuvant concurrent chemoradiation in patients with locally advanced breast cancer (LABC). We now report disease-free survival (DFS) and overall survival (OS) in the context of pathologic response. 105 LABC patients (White 46%, Non-White 54%) were treated with paclitaxel (30 mg/m² intravenously twice a week) for 10-12 weeks. Daily radiotherapy was delivered to breast, axillary, and supraclavicular lymph nodes during weeks 2-7 of paclitaxel treatment, at 1.8 Gy per fraction to a total dose of 45 Gy with a tumor boost of 14 Gy at 2 Gy/fraction. Pathological complete response (pCR) was defined as the absence of invasive cancer in breast and lymph nodes and pathological partial response (pPR) as the persistence of <10 microscopic foci of invasive carcinoma in breast or lymph nodes. Pathologic response (pCR and pPR) after neoadjuvant chemoradiation was achieved in 36/105 patients (34%) and was associated with significantly better DFS and OS. Pathological responders had a lower risk of recurrence or death (HR = 0.35, P = 0.01) and a longer OS (HR = 4.27, P = 0.01) compared with non-responders. Median DFS and OS were 57 and 84 months for non-responders, respectively, and have not yet been reached for responders. Importantly, pathologic response was achieved in 54% of patients with HR negative tumors (26/48). In conclusion, pathologic response to concurrent paclitaxel-radiation translated into superior DFS and OS. Half of the patients with HR negative tumors achieved a pathologic response.
Collapse
|
28
|
Ling Y, Fan L, Dong C, Zhu J, Liu Y, Ni Y, Zhu C, Zhang C. Combined influence of adjuvant therapy and interval after surgery on peripheral CD4(+) T lymphocytes in patients with esophageal squamous cell carcinoma. Exp Ther Med 2010; 1:113-120. [PMID: 23136603 DOI: 10.3892/etm_00000020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 10/26/2009] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to investigate possible differences in cellular immunity between chemo- and/or radiotherapy groups during a long interval after surgery in esophageal squamous cell carcinoma (ESCC) patients. Cellular immunity was assessed as peripheral lymphocyte subsets in response to chemotherapy (CT), radiotherapy (RT) and CT+RT by flow cytometric analysis. There were 139 blood samples obtained at different time points relative to surgery from 73 patients with ESCC. The changes in the absolute and relative proportions of lymphocyte phenotypes were significant among the adjuvant therapy groups. There were significant differences in the absolute counts of CD4(+) and CD8(+) T cells among the interval groups, and a lower CD4/CD8 ratio was found in patients following a prolonged interval. RT alone had a profound effect on the absolute counts of CD3(+), CD4(+) and CD8(+) T cells compared with the other groups. CD4(+) T cells exhibited a decreasing trend during a long interval, leading to a prolonged T-cell imbalance after surgery. Univariate analysis revealed that the interaction of the type of adjuvant therapy and the interval after surgery was correlated only with the percentage of CD4(+) T cells. The percentage of CD4(+) T cells can be used as an indicator of the cellular immunity after surgery in ESCC patients. However, natural killer cells consistently remained suppressed in ESCC patients following adjuvant therapy after surgery. These findings confirm an interaction between adjuvant therapy and the interval after surgery on peripheral CD4(+) T cells, and implies that adjuvant therapy may have selective influence on the cellular immunity of ESCC patients after surgery.
Collapse
Affiliation(s)
- Yang Ling
- Laboratory of Clinical Oncology, Changzhou Tumor Hospital, Medical College of Soochow University, Jiangsu
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Radiotherapy is a local treatment modality employed in breast cancer to reduce local recurrence following surgery. The observed association of optimal local control with improved survival was not expected in a disease characterized by early systemic spread. The underlying mechanisms whereby the application of ionizing radiation to the primary tumor site can have systemic effects remain unclear and are the subject of much debate. In the present article we discuss the hypothesis that radiotherapy has unique biological effects and that, in addition to killing residual neoplastic cells after surgery is performed, it might favorably alter the microenvironment at the primary tumor site during the process of wound healing and the development of antitumor immune responses.
Collapse
Affiliation(s)
- Silvia C Formenti
- Department of Radiation Oncology, New York University School of Medicine and NYU Cancer Institute, 160 East 34th Street, New York, NY 10016, USA.
| | | |
Collapse
|
30
|
Campana L, Bosurgi L, Rovere-Querini P. HMGB1: a two-headed signal regulating tumor progression and immunity. Curr Opin Immunol 2008; 20:518-23. [PMID: 18599281 DOI: 10.1016/j.coi.2008.04.012] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Revised: 04/28/2008] [Accepted: 04/29/2008] [Indexed: 01/28/2023]
Abstract
Cells of the innate immune system sense tissue damage recognizing in the extracellular environment bona fide intracellular moieties, like high mobility group box 1 (HMGB1). In the case of tumors, HMGB1 recognition has a paradoxical dual effect: it promotes tumor neoangiogenesis and triggers protective anti-neoplastic T-cell responses. Recent advances in the study of HMGB1 have identified candidate molecular mechanisms underlying these apparently contrasting outcomes. A surprising role for innate receptors, including toll like receptor 4 (TLR4), in the response to conventional cancer radio and chemotherapy has also recently emerged, providing new insight into the mechanisms by which these treatments actually work.
Collapse
Affiliation(s)
- Lara Campana
- Laboratory of Clinical Immunology, San Raffaele Scientific Institute, DIBIT 3A1, Via Olgettina 58, 20132 Milano, Italy
| | | | | |
Collapse
|