1
|
Quan L, Liu J, Wang Y, Yang F, Yang Z, Ju J, Shuai Y, Wei T, Yue J, Wang X, Meng J, Yuan P. Exploring risk factors for endocrine-related immune-related adverse events: Insights from meta-analysis and Mendelian randomization. Hum Vaccin Immunother 2024; 20:2410557. [PMID: 39377304 PMCID: PMC11469449 DOI: 10.1080/21645515.2024.2410557] [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: 06/24/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024] Open
Abstract
This study utilized meta-analysis and Mendelian randomization (MR) to identify risk factors for endocrine-related immune-related adverse events (EirAEs) and to ascertain whether EirAEs confer better prognosis of immunotherapy. The meta-analysis identified several risk factors for EirAEs, including elevated baseline TSH (OR = 1.30, 95% CI 1.10-1.53), positive TgAb (OR = 14.23, p < .001), positive TPOAb (OR = 3.75, p < .001), prior thyroid-related medical history (OR = 4.19), increased BMI (OR = 1.11), combination immune checkpoint inhibitors (ICIs) therapy with targeted treatment (OR = 2.71, 95% CI 2.11-3.47), and dual ICI therapy (OR = 3.26, 95% CI 2.22-4.79). MR analysis further supported causalities between extreme BMI, hypothyroidism, and irAEs from a genetic perspective. In addition, cancer patients who experienced EirAEs exhibited significantly prolonged PFS (HR = 0.84, 95% CI 0.73-0.97) and OS (HR = 0.59, 95% CI 0.45-0.76) compared to those without. These findings provide valuable insights for clinical decision-making among healthcare professionals and offer direction for future research in this field.
Collapse
Affiliation(s)
- Liuliu Quan
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinsong Liu
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxin Wang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fan Yang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Zixuan Yang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Ju
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - You Shuai
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tong Wei
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian Yue
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Wang
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaqi Meng
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Peng Yuan
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Hou Y, Zhang F, Zong J, Li T, Gan W, Lv S, Yan Z, Zeng Z, Yang L, Zhou M, Zhao W, Yang M. Integrated analysis reveals a novel 5-fluorouracil resistance-based prognostic signature with promising implications for predicting the efficacy of chemotherapy and immunotherapy in patients with colorectal cancer. Apoptosis 2024; 29:1126-1144. [PMID: 38824480 DOI: 10.1007/s10495-024-01981-2] [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] [Accepted: 05/14/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND 5-Fluorouracil (5-FU) has been used as a standard first-line treatment for colorectal cancer (CRC) patients. Although 5-FU-based chemotherapy and immune checkpoint blockade (ICB) have achieved success in treating CRC, drug resistance and low response rates remain substantial limitations. Thus, it is necessary to construct a 5-FU resistance-related signature (5-FRSig) to predict patient prognosis and identify ideal patients for chemotherapy and immunotherapy. METHODS Using bulk and single-cell RNA sequencing data, we established and validated a novel 5-FRSig model using stepwise regression and multiple CRC cohorts and evaluated its associations with the prognosis, clinical features, immune status, immunotherapy, neoadjuvant therapy, and drug sensitivity of CRC patients through various bioinformatics algorithms. Unsupervised consensus clustering was performed to categorize the 5-FU resistance-related molecular subtypes of CRC. The expression levels of 5-FRSig, immune checkpoints, and immunoregulators were determined using quantitative real-time polymerase chain reaction (RT‒qPCR). Potential small-molecule agents were identified via Connectivity Map (CMap) and molecular docking. RESULTS The 5-FRSig and cluster were confirmed as independent prognostic factors in CRC, as patients in the low-risk group and Cluster 1 had a better prognosis. Notably, 5-FRSig was significantly associated with 5-FU sensitivity, chemotherapy response, immune cell infiltration, immunoreactivity phenotype, immunotherapy efficiency, and drug selection. We predicted 10 potential compounds that bind to the core targets of 5-FRSig with the highest affinity. CONCLUSION We developed a valid 5-FRSig to predict the prognosis, chemotherapeutic response, and immune status of CRC patients, thus optimizing the therapeutic benefits of chemotherapy combined with immunotherapy, which can facilitate the development of personalized treatments and novel molecular targeted therapies for patients with CRC.
Collapse
Affiliation(s)
- Yufang Hou
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 2 Nanwei Road, Beijing, 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Fang Zhang
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 2 Nanwei Road, Beijing, 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jinbao Zong
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
- Qingdao Hospital of Traditional Chinese Medicine, The affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, 266033, China
| | - Tiegang Li
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 2 Nanwei Road, Beijing, 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wenqiang Gan
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 2 Nanwei Road, Beijing, 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Silin Lv
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 2 Nanwei Road, Beijing, 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zheng Yan
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 2 Nanwei Road, Beijing, 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zifan Zeng
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 2 Nanwei Road, Beijing, 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Liu Yang
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 2 Nanwei Road, Beijing, 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Mingxuan Zhou
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 2 Nanwei Road, Beijing, 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wenyi Zhao
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 2 Nanwei Road, Beijing, 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Min Yang
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 2 Nanwei Road, Beijing, 100050, China.
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
3
|
Socinski MA, Jotte RM, Cappuzzo F, Nishio M, Mok TSK, Reck M, Finley GG, Kaul MD, Yu W, Paranthaman N, Bāra I, West HJ. Association of Immune-Related Adverse Events With Efficacy of Atezolizumab in Patients With Non-Small Cell Lung Cancer: Pooled Analyses of the Phase 3 IMpower130, IMpower132, and IMpower150 Randomized Clinical Trials. JAMA Oncol 2023; 9:527-535. [PMID: 36795388 PMCID: PMC9936386 DOI: 10.1001/jamaoncol.2022.7711] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/29/2022] [Indexed: 02/17/2023]
Abstract
Importance Immune-related adverse events (irAEs) arising from immune checkpoint inhibitor (ICI) cancer therapy may potentially predict improved outcomes. Objective To evaluate the association between irAEs and atezolizumab efficacy in patients with advanced non-small cell lung cancer (NSCLC) using pooled data from 3 phase 3 ICI studies. Design, Setting, and Participants IMpower130, IMpower132, and IMpower150 were phase 3, multicenter, open-label, randomized clinical trials to evaluate the efficacy and safety of chemoimmunotherapy combinations involving atezolizumab. Participants were chemotherapy-naive adults with stage IV nonsquamous NSCLC. These post hoc analyses were conducted during February 2022. Interventions Eligible patients were randomly assigned 2:1 to receive atezolizumab with carboplatin plus nab-paclitaxel, or chemotherapy alone (IMpower130); 1:1 to receive atezolizumab with carboplatin or cisplatin plus pemetrexed, or chemotherapy alone (IMpower132); and 1:1:1 to receive atezolizumab plus bevacizumab plus carboplatin and paclitaxel, atezolizumab plus carboplatin and paclitaxel, or bevacizumab plus carboplatin and paclitaxel (IMpower150). Main Outcomes and Measures Pooled data from IMpower130 (cutoff: March 15, 2018), IMpower132 (cutoff: May 22, 2018), and IMpower150 (cutoff: September 13, 2019) were analyzed by treatment (atezolizumab-containing vs control), irAE status (with vs without), and highest irAE grade (1-2 vs 3-5). To account for immortal bias, a time-dependent Cox model and landmark analyses of irAE occurrence at 1, 3, 6, and 12 months from baseline were used to estimate the hazard ratio (HR) of overall survival (OS). Results Of 2503 randomized patients, 1577 were in the atezolizumab-containing arm and 926 were in the control arm. The mean (SD) age of patients was 63.1 (9.4) years and 63.0 (9.3) years, and 950 (60.2%) and 569 (61.4%) were male, respectively, in the atezolizumab arm and the control arm. Baseline characteristics were generally balanced between patients with irAEs (atezolizumab, n = 753; control, n = 289) and without (atezolizumab, n = 824; control, n = 637). In the atezolizumab arm, OS HRs (95% CI) in patients with grade 1 to 2 irAEs and grade 3 to 5 irAEs (each vs those without irAEs) in the 1-, 3-, 6-, and 12-month subgroups were 0.78 (0.65-0.94) and 1.25 (0.90-1.72), 0.74 (0.63-0.87) and 1.23 (0.93-1.64), 0.77 (0.65-0.90) and 1.1 (0.81-1.42), and 0.72 (0.59-0.89) and 0.87 (0.61-1.25), respectively. Conclusions and Relevance In this pooled analysis of 3 randomized clinical trials, longer OS was observed in patients with vs without mild to moderate irAEs in both arms and across landmarks. These findings further support the use of first-line atezolizumab-containing regimens for advanced nonsquamous NSCLC. Trial Registration ClinicalTrials.gov Identifiers: NCT02367781, NCT02657434, and NCT02366143.
Collapse
Affiliation(s)
| | - Robert M. Jotte
- Rocky Mountain Cancer Centers, Denver, Colorado
- US Oncology, Houston, Texas
| | | | - Makoto Nishio
- The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tony S. K. Mok
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong
| | - Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, German Center for Lung Research, Grosshansdorf, Germany
| | - Gene G. Finley
- Allegheny Health Network Cancer Institute, Pittsburgh, Pennsylvania
| | | | - Wei Yu
- Genentech, Inc, South San Francisco, California
| | | | - Ilze Bāra
- Genentech, Inc, South San Francisco, California
| | - Howard J. West
- City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
4
|
Zhang P, Li Q, Zhang Y, Wang Q, Yan J, Shen A, Hu B. Identification of a Novel Gene Signature with DDR and EMT Difunctionalities for Predicting Prognosis, Immune Activity, and Drug Response in Breast Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1221. [PMID: 36673982 PMCID: PMC9859620 DOI: 10.3390/ijerph20021221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/02/2023] [Accepted: 01/07/2023] [Indexed: 06/17/2023]
Abstract
Breast cancer, with an overall poor clinical prognosis, is one of the most heterogeneous cancers. DNA damage repair (DDR) and epithelial-mesenchymal transition (EMT) have been identified to be associated with cancer's progression. Our study aimed to explore whether genes with both functions play a more crucial role in the prognosis, immune, and therapy response of breast cancer patients. Based on the Cancer Genome Atlas (TCGA) cancer database, we used LASSO regression analysis to identify the six prognostic-related genes with both DDR and EMT functions, including TP63, YWHAZ, BRCA1, CCND2, YWHAG, and HIPK2. Based on the six genes, we defined the risk scores of the patients and reasonably analyzed the overall survival rate between the patients with the different risk scores. We found that overall survival in higher-risk-score patients was lower than in lower-risk-score patients. Subsequently, further GO and KEGG analyses for patients revealed that the levels of immune infiltration varied for patients with high and low risk scores, and the high-risk-score patients had lower immune infiltration's levels and were insensitive to treatment with chemotherapeutic agents. Furthermore, the Gene Expression Omnibus (GEO) database validated our findings. Our data suggest that TP63, YWHAZ, BRCA1, CCND2, YWHAG, and HIPK2 can be potential genetic markers of prognostic assessment, immune infiltration and chemotherapeutic drug sensitivity in breast cancer patients.
Collapse
Affiliation(s)
- Pan Zhang
- Department of Radiation Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Watershed Science and Health of Zhejiang Province, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Quan Li
- Department of Radiation Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Watershed Science and Health of Zhejiang Province, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Yuni Zhang
- Department of Radiation Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Watershed Science and Health of Zhejiang Province, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Qianqian Wang
- Department of Radiation Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Watershed Science and Health of Zhejiang Province, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Junfang Yan
- Department of Radiation Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Watershed Science and Health of Zhejiang Province, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Aihua Shen
- Department of Radiation Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Watershed Science and Health of Zhejiang Province, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Burong Hu
- Department of Radiation Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Watershed Science and Health of Zhejiang Province, School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
5
|
de Santana Cerqueira N, Vitória LA, da Silva VP, Rodriguez TT, Cangussu MCT, Ramalho LMP. Expression and degranulation of mast cells in laser photobiomodulated mucositis chemo-induced: pilot study in hamsters. Lasers Med Sci 2023; 38:31. [PMID: 36595054 DOI: 10.1007/s10103-022-03698-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/20/2022] [Indexed: 01/04/2023]
Abstract
This study aimed to evaluate the influence of laser photobiomodulation on the expression and degranulation of mast cells in chemo-induced oral mucositis (OM) lesions in hamsters. Twelve adult male Syrian hamsters (Mesocricetus auratus), golden lineage, were submitted to OM induction. They were divided into three groups: control-OM without treatment (C), OM treated with red laser (RL), OM treated with infrared laser (IL) and analyzed in the experimental time of 7 days. Three and 4 days after the intraperitoneal injection of the chemotherapy drug fluorouracil, the OM lesions were induced by making grooves in the right cheek pouch. Immediately after chemoinduction, the hamsters were submitted to photobiomodulation every 48 h for 7 days. The specimens were processed and stained using the hematoxylin-eosin and toluidine blue techniques. There was a predominance of mild chronic inflammation in the experimental groups and a greater persistence of neutrophils in the control group (C), although not statistically significant. The group irradiated with red laser (RL) had the highest mean mast cell expression (38.28 ± 19.05) (p < 0.001). As for the degranulation activity in mast cells, the control group (C) showed a greater number of fields with more than 50% of degranulated cells, presenting statistical significance when comparing it with the RL (p < 0.009) and IL (p = 0.036) group. It can be concluded that photobiomodulation, at both wavelengths, decreased mast cell degranulation, accelerating the inflammatory process. The use of infrared laser provided, in addition to less degranulation, the quantitative reduction of mast cells.
Collapse
Affiliation(s)
- Naiadja de Santana Cerqueira
- Postgraduate Program in Dentistry and Health, Faculty of Dentistry, Federal University of Bahia, Salvador, Bahia, Brazil
| | | | | | - Tânia Tavares Rodriguez
- Department of Bioregulation, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | | | - Luciana Maria Pedreira Ramalho
- Department of Propaedeutics and Integrated Clinic, Faculty of Dentistry, Federal University of Bahia, Araújo Pinho Avenue, no. 62, Canela, 9th floor, Salvador, Bahia, 40301-155, Brazil.
| |
Collapse
|
6
|
Ni J, Si X, Wang H, Zhang X, Zhang L. Camrelizumab plus platinum-irinotecan followed by maintenance camrelizumab plus apatinib in untreated extensive-stage small-cell lung cancer: a nonrandomized clinical trial. Front Immunol 2023; 14:1168879. [PMID: 37114054 PMCID: PMC10126331 DOI: 10.3389/fimmu.2023.1168879] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
Background Programmed cell death-ligand 1 (PD-L1) inhibitors plus chemotherapy have made substantial progress in extensive-stage small-cell lung cancer (ES-SCLC), but the survival benefit is still limited. This study aimed to evaluate the preliminary efficacy and safety of camrelizumab plus platinum-irinotecan (IP/IC) followed by maintenance camrelizumab plus apatinib in patients with untreated ES-SCLC. Methods In this non-randomized clinical trial (NCT04453930), eligible patients with untreated ES-SCLC received 4-6 cycles of camrelizumab plus IP/IC, followed by maintenance with camrelizumab plus apatinib until disease progression or unmanageable toxicity. The primary endpoint was progression-free survival (PFS). Patients who received PD-L1 inhibitors (atezolizumab or durvalumab) plus platinum-etoposide (EP/EC) were selected as the historical control. Results Nineteen patients received IP/IC plus camrelizumab and 34 patients received EP/EC plus PD-L1 inhibitor. At a median follow-up time of 12.1 months, the median PFS was 10.25 months (95% CI: 9.40-NA) in the IP/IC plus camrelizumab group and 7.10 months (95% CI 5.79-8.40) in the EP/EC plus PD-L1 inhibitor group, respectively (HR=0.58, 95% CI 0.42-0.81). The objective response rate of IP/IC plus camrelizumab and EP/EC plus PD-L1 inhibitor was 89.6% and 82.4%, respectively. The most common treatment-related adverse events in the IP/IC plus camrelizumab group was neutropenia, followed by reactive cutaneous capillary endothelial proliferation (RCCEP) and diarrhea. The occurrence of immune-related adverse event was found to be associated with a prolonged PFS (HR=4.64, 95% CI 1.92-11.18). Conclusions IP/IC plus camrelizumab followed by maintenance camrelizumab plus apatinib showed preliminary efficacy and acceptable safety profile in patients with untreated ES-SCLC.
Collapse
Affiliation(s)
- Jun Ni
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences & Peking Union Medical College, Beijing, China
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Si
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences & Peking Union Medical College, Beijing, China
| | - Hanping Wang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences & Peking Union Medical College, Beijing, China
| | - Xiaotong Zhang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences & Peking Union Medical College, Beijing, China
- *Correspondence: Li Zhang, ; Xiaotong Zhang,
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences & Peking Union Medical College, Beijing, China
- *Correspondence: Li Zhang, ; Xiaotong Zhang,
| |
Collapse
|
7
|
Zhang SQ, Pan SM, Lai SZ, Situ HJ, Liu J, Dai WJ, Liang SX, Zhou LQ, Lu QQ, Ke PF, Zhang F, Chen HB, Li JC. Novel Plasma Proteomic Biomarkers for Early Identification of Induction Chemotherapy Beneficiaries in Locoregionally Advanced Nasopharyngeal Carcinoma. Front Oncol 2022; 12:889516. [PMID: 35847896 PMCID: PMC9279567 DOI: 10.3389/fonc.2022.889516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/02/2022] [Indexed: 02/05/2023] Open
Abstract
Background Induction chemotherapy (IC) can alleviate locoregionally advanced nasopharyngeal carcinoma (LA-NPC), but effectiveness differs between patients, toxicity is problematic, and effective blood-based IC efficacy predictors are lacking. Here, we aimed to identify biomarkers for early identification of IC beneficiaries. Methods Sixty-four pairs of matched plasma samples collected before and after IC from LA-NPC patients including 34 responders and 30 non-responders, as well as 50 plasma samples of healthy individuals, were tested using data-independent acquisition mass spectrometry. The proteins associated with clinical traits or IC benefits were investigated by weighted gene co-expression network analysis (WGCNA) and soft cluster analysis. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional annotations were performed to determine the potential function of the identified proteins. The area under the receiver operating characteristic curve (AUC) was used to evaluate the performance of candidate biomarkers in predicting IC beneficiaries. Results Compared with healthy individuals, 1027 differentially expressed proteins (DEPs) were found in the plasma of LA-NPC patients. Based on feedback from IC outcomes, 463 DEPs were identified in the pre-IC plasma between responders and non-responders. A total of 1212 DEPs represented the proteomic changes before and after IC in responders, while 276 DEPs were identified in post-IC plasma between responders and non-responders. WGCNA identified nine protein co-expression modules correlated with clinical traits. Soft cluster analysis identified four IC benefits-related protein clusters. Functional enrichment analysis showed that these proteins may play a role in IC via immunity, complement, coagulation, glycosaminoglycan and serine. Four proteins differentially expressed in all group comparisons, paraoxonase/arylesterase 1 (PON1), insulin-like growth factor-binding protein 3 (IGFBP-3), rheumatoid factor D5 light chain (v-kappa-3) and RNA helicase (DDX55), were associated with clinical traits or IC benefits. A four-protein model accurately identified potential IC beneficiaries (AUC=0.95) while diagnosing LA-NPC (AUC=0.92), and the prediction performance was verified using the models to confirm the effective IC (AUC=0.97) and evaluate IC outcome (AUC=0.94). Conclusion The plasma protein profiles among IC responders and non-responders were different. PON1, IGFBP3, v-kappa-3 and DDX55 could serve as potential biomarkers for early identification of IC beneficiaries for individualised treatment of LA-NPC.
Collapse
Affiliation(s)
- Shan-Qiang Zhang
- Medical Research Center, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Su-Ming Pan
- Department of Radiation Oncology, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Shu-Zhen Lai
- Department of Radiation Oncology, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Hui-Jing Situ
- Department of Radiation Oncology, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Jun Liu
- Medical Research Center, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Wen-Jie Dai
- Medical Research Center, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Si-Xian Liang
- Medical Research Center, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Li-Qing Zhou
- Medical Research Center, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Qi-Qi Lu
- Medical Research Center, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Pei-Feng Ke
- Medical Research Center, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Fan Zhang
- Medical Research Center, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Hai-Bin Chen
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
| | - Ji-Cheng Li
- Medical Research Center, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
- Institute of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
8
|
Konopka K, Frączek P, Lubaś M, Micek A, Kwinta Ł, Streb J, Potocki P, Wysocki PJ. Reduction of Cancer-Induced Thrombocytosis as a Biomarker of Improved Outcomes in Advanced Gastric Cancer. J Clin Med 2022; 11:jcm11051213. [PMID: 35268305 PMCID: PMC8911022 DOI: 10.3390/jcm11051213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Interplay between non-specific inflammatory reaction and tumor microenvironment in gastric cancer (GC) can be measured indirectly by assessing fluctuations in concentration of platelets. Cytotoxic chemotherapy affects these morphotic elements directly by inducing myelosuppression. It was hypothesized that chemotherapy not only directly affects malignant cells, but also through immunomodulation related to myelosuppression. METHODS Metastatic GC patients (N: 155) treated with chemotherapy +/- trastuzumab were enrolled in this retrospective study. Platelet pretreatment concentration (PLT-count) and the deepest level of platelet reduction, as well as other inflammatory and general confounders were collected in the first 12 weeks of treatment (PLT-red). Martingale residuals were used to visualize the relationship between PLT-count, PLT-red, and overall survival (OS). Multiple multivariate Cox regression models were built to assess the impact of platelet reduction on OS and progression-free survival (PFS). RESULTS Reduction of PLT (PLT-red) to 60% of baseline concentration was associated with improved survival rates (HR = 0.60, p = 0.026 for OS and HR 0.56, p = 0.015 for PFS). Cross-classification into four groups based on PLT-count (high vs low) and PLT-red (high vs low) showed significantly worse survival rates in both high PLT-count (HR = 3.60, p = 0.007 for OS and HR = 2.97, p = 0.024 for PFS) and low PLT-count (HR = 1.75, p = 0.035 for OS and HR = 1.80, p = 0.028 for PFS) patients with insufficient platelets reduction. CONCLUSION Thrombocytosis reduction represents a novel, clinically important, prognostic factor for OS and PFS in patients with stage IV GC.
Collapse
Affiliation(s)
- Kamil Konopka
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland
- Correspondence:
| | - Paulina Frączek
- Department of Medical Oncology, University Hospital in Cracow, 30-688 Cracow, Poland
| | - Maciej Lubaś
- Department of Medical Oncology, University Hospital in Cracow, 30-688 Cracow, Poland
| | - Agnieszka Micek
- Department of Nursing Management and Epidemiology Nursing, Jagiellonian University Medical College, 31-007 Cracow, Poland
| | - Łukasz Kwinta
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland
| | - Joanna Streb
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland
| | - Paweł Potocki
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland
| | - Piotr J. Wysocki
- Department of Oncology, Jagiellonian University Medical College, 31-007 Cracow, Poland
| |
Collapse
|
9
|
Fathi M, Vakili K, Jazi K, Sadeghi MA, Hajiesmaeili M, Mohamadkhani A, Rezaei-Tavirani M, Tavasol A. Challenges of cancer immunotherapy and chemotherapy during the COVID-19 pandemic. TUMORI JOURNAL 2021; 108:407-419. [PMID: 34918602 DOI: 10.1177/03008916211063939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
People at high risk of morbidity and mortality from coronavirus disease 2019 (COVID-19), including patients dealing with malignancies and patients on immunosuppressive anticancer therapies, need to be followed carefully as the pandemic continues. Challenges in continuing cancer management and patient monitoring are of concern given the importance of timing in cancer therapy. Alternative treatment decisions and priorities are also important considerations. The efficacy and safety of various cancer treatments in patients with COVID-19 are other important considerations. In this systematic review, we summarize the potential risks and benefits of cancer treatments applied to patients with COVID-19 and malignant tumors. Using the PubMed and Scopus databases, we reviewed studies involving cancer therapy and COVID-19 to address the recent discoveries and related challenges of cancer therapy in patients with COVID-19 and cancer.
Collapse
Affiliation(s)
- Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Jazi
- Student Research Committee, Faculty of Medicine, Medical University of Qom, Qom, Iran
| | | | - Mohammadreza Hajiesmaeili
- Critical Care Quality Improvement Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ashraf Mohamadkhani
- Digestive Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arian Tavasol
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Polansky H, Goral B. How an increase in the copy number of HSV-1 during latency can cause Alzheimer's disease: the viral and cellular dynamics according to the microcompetition model. J Neurovirol 2021; 27:895-916. [PMID: 34635992 DOI: 10.1007/s13365-021-01012-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 04/28/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022]
Abstract
Numerous studies observed a link between the herpes smplex virus-1 (HSV-1) and Alzheimer's disease. However, the exact viral and cellular dynamics that lead from an HSV-1 infection to Alzheimer's disease are unknown. In this paper, we use the microcompetition model to formulate these dynamics by connecting seemingly unconnected observations reported in the literature. We concentrate on four pathologies characteristic of Alzheimer's disease. First, we explain how an increase in the copy number of HSV-1 during latency can decrease the expression of BECN1/Beclin1, the degradative trafficking protein, which, in turn, can cause a dysregulation of autophagy and Alzheimer's disease. Second, we show how an increase in the copy number of the latent HSV-1 can decrease the expression of many genes important for mitochondrial genome metabolism, respiratory chain, and homeostasis, which can lead to oxidative stress and neuronal damage, resulting in Alzheimer's disease. Third, we describe how an increase in this copy number can reduce the concentration of the NMDA receptor subunits NR1 and NR2b (Grin1 and Grin2b genes), and brain derived neurotrophic factor (BDNF), which can cause an impaired synaptic plasticity, Aβ accumulation and eventually Alzheimer's disease. Finally, we show how an increase in the copy number of HSV-1 in neural stem/progenitor cells in the hippocampus during the latent phase can lead to an abnormal quantity and quality of neurogenesis, and the clinical presentation of Alzheimer's disease. Since the current understanding of the dynamics and homeostasis of the HSV-1 reservoir during latency is limited, the proposed model represents only a first step towards a complete understanding of the relationship between the copy number of HSV-1 during latency and Alzheimer's disease.
Collapse
Affiliation(s)
- Hanan Polansky
- The Center for the Biology of Chronic Disease (CBCD), 3 Germay Dr, Wilmington, DE, 19804, USA.
| | - Benjamin Goral
- The Center for the Biology of Chronic Disease (CBCD), 3 Germay Dr, Wilmington, DE, 19804, USA
| |
Collapse
|
11
|
Glorieux C, Xia X, You X, Wang Z, Han Y, Yang J, Noppe G, Meester CD, Ling J, Robert A, Zhang H, Li SP, Wang H, Chiao PJ, Zhang L, Li X, Huang P. Cisplatin and gemcitabine exert opposite effects on immunotherapy with PD-1 antibody in K-ras-driven cancer. J Adv Res 2021; 40:109-124. [PMID: 36100320 PMCID: PMC9481954 DOI: 10.1016/j.jare.2021.12.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/01/2021] [Accepted: 12/14/2021] [Indexed: 12/17/2022] Open
Abstract
Two common chemotherapeutic drugs, cisplatin and gemcitabine, exert opposite effect on the efficacy of PD-1 antibody in K-ras-driven cancers. Gemcitabine antagonizes PD-1Ab due to its inhibition on T cell infiltration in tumor tissues. Combination PD-1Ab and cisplatin leads to complete tumor eradication in vivo due to activation of the cGAS-mediated immune response. The impact of drugs on T cell functions should be considered as a critical factor in selecting drugs for immunochemotherapy to achieve optimal therapeutic outcome.
Introduction Immunochemotherapy using PD-1/PD-L1 antibodies in combination with chemotherapeutic agents has become a mainstream treatment for cancer patients, but it remains unclear which drug combinations would produce best therapeutic outcome. Objectives The purpose of this study was to investigate two common chemotherapeutic drugs, gemcitabine and cisplatin, for their impacts on the therapeutic efficacy of PD-1 antibody in K-ras-driven cancers known to overexpress PD-L1. Methods Both in vitro assays and syngeneic mouse tumor models were used in this study. Biochemical and molecular assays were used to determine the effects of drugs on T cell functions in cell culture models and in mouse/human tumor tissues. Allograft tumor models with K-ras mutation were used to investigate the combination effect of gemcitabine or cisplatin with immunotherapy. Data of lung cancer patients with K-ras mutation treated with cisplatin and toripalimab were analyzed to evaluate the clinical relevance of the lab findings. Results Cisplatin and gemcitabine unexpectedly exert opposite effect on the therapeutic activity of PD-1 antibody in vivo. Gemcitabine antagonizes the therapeutic effect of PD-1 antibody due to its significant inhibition on CD8+ T cell infiltration, which was observed both in mouse tumor allografts and in human pancreatic cancer tissues. In contrast, cisplatin shows synergistic activity with PD-1 antibody by activation of CD8+ T cells through the DNA damage-mediated cGAS-STING sensing mechanism, leading to increase of T cell infiltration and secretion of antitumor cytokines. Clinical data show that a combination of cisplatin with PD-1 antibody toripalimab could be effective in advanced lung cancer patients with K-ras mutation who failed prior therapies. Conclusions Our study shows that a key factor in selecting chemotherapeutic agents for immunochemotherapy is the drug’s impact on T cell functions, and that cisplatin-based chemotherapy is an excellent choice for combination with immune checkpoint antibody to achieve favorable clinical outcome.
Collapse
|
12
|
Huang W, Chen JJ, Xing R, Zeng YC. Combination therapy: Future directions of immunotherapy in small cell lung cancer. Transl Oncol 2021; 14:100889. [PMID: 33065386 PMCID: PMC7567053 DOI: 10.1016/j.tranon.2020.100889] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/31/2022] Open
Abstract
Small cell lung cancer (SCLC), an aggressive and devastating malignancy, is characterized by rapid growth and early metastasis. Although most patients respond to first-line chemotherapy, the majority of patients rapidly relapse and have a relatively poor prognosis. Fortunately, immunotherapy, mainly including antibodies that target the cytotoxic T lymphocyte antigen-4 (CTLA-4), checkpoints programmed death-1 (PD-1), and programmed death-ligand 1 (PD-L1) to block immune regulatory checkpoints on tumor cells, immune cells, fibroblasts cells and endothelial cells, has achieved the milestone in several solid tumors, such as melanoma and non-small-cell lung carcinomas (NSCLC). In recent years, immunotherapy has made progress in the treatment of patients with SCLC, while its response rate is relatively low to monotherapy. Interestingly, the combination of immunotherapy with other therapy, such as chemotherapy, radiotherapy, and targeted therapy, preliminarily achieve greater therapeutic effects for treating SCLC. Combining different immunotherapy drugs may act synergistically because of the complementary effects of the two immune checkpoint pathways (CTLA-4 and PD-1/PD-L1 pathways). The incorporation of chemoradiotherapy in immunotherapy may augment antitumor immune responses because chemoradiotherapy can enhance tumor cell immunogenicity by rapidly inducing tumor lysis and releasing tumor antigens. In addition, since immunotherapy drugs and the molecular targets drugs act on different targets and cells, the combination of these drugs may achieve greater therapeutic effects in the treatment of SCLC. In this review, we focused on the completed and ongoing trials of the combination therapy for immunotherapy of SCLC to find out the rational combination strategies which may improve the outcomes for SCLC.
Collapse
Affiliation(s)
- Wei Huang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China; Department of Clinical Oncology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang 110022, China
| | - Jia-Jia Chen
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang 110022, China
| | - Rui Xing
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang 110022, China
| | - Yue-Can Zeng
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang 110022, China; Department of Medical Oncology, Cancer Center, The Second Affiliated Hospital of Hainan Medical University, 368 Yehai Road, Haikou 571199, China.
| |
Collapse
|
13
|
Sui S, An X, Xu C, Li Z, Hua Y, Huang G, Sui S, Long Q, Sui Y, Xiong Y, Ntim M, Guo W, Chen M, Li M, Xiao X, Deng W. An immune cell infiltration-based immune score model predicts prognosis and chemotherapy effects in breast cancer. Am J Cancer Res 2020; 10:11938-11949. [PMID: 33204321 PMCID: PMC7667685 DOI: 10.7150/thno.49451] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 09/18/2020] [Indexed: 01/01/2023] Open
Abstract
Background: Immune cells have essential auxiliary functions and influence clinical outcomes in cancer, with high immune infiltration being associated with improved clinical outcomes and better response to treatment in breast cancer (BC). However, studies to date have not fully considered the tumor-infiltrating immune cell (TIIC) landscape in tumors. This study investigated potential biomarkers based on TIICs to improve prognosis and treatment effect in BC. Results: We enrolled 5112 patients for analysis and used cell type identification by estimating relative subsets of RNA transcripts (CIBERSORT), a new computational algorithm, to quantify 22 TIICs in primary BC. From the results of univariate Cox regression, 12 immune cells were determined to be significantly related to the overall survival (OS) of BC patients. Furthermore, least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression analyses were applied to construct an immune prognostic model based on six potential biomarkers. By dividing patients into low- and high-risk groups, a significant distinction in OS was found in the training cohort, with 20-year survival rates of 42.6% and 26.3%, respectively. Applying a similar protocol to validation and test cohorts, we found that OS was significantly shorter in the high-risk group than in the low-risk group, regardless of the molecular subtype of BC. Using the immune score model to predict the effect of BC patients to chemotherapy, the survival advantage for the low-risk group was evident among those who received chemotherapy, regardless of the chemotherapy regimen. In evaluating the predictive value of the nomogram, a decision curve showed better predictive accuracy than the standard tumor-node-metastasis (TNM) staging system. Conclusion: The immune cell infiltration-based immune score model can be effectively and efficiently used to predict the prognosis of BC patients as well as the effect of chemotherapy.
Collapse
|
14
|
Li C, Donninger H, Eaton J, Yaddanapudi K. Regulatory Role of Immune Cell-Derived Extracellular Vesicles in Cancer: The Message Is in the Envelope. Front Immunol 2020; 11:1525. [PMID: 32765528 PMCID: PMC7378739 DOI: 10.3389/fimmu.2020.01525] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/09/2020] [Indexed: 12/28/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogenous group of membrane-surrounded structures. Besides serving as a harbor for the unwanted material exocytosed by cells, EVs play a critical role in conveying intact protein, genetic, and lipid contents that are important for intercellular communication. EVs, broadly comprised of microvesicles and exosomes, are released to the extracellular environment from nearly all cells either via shedding from the plasma membrane or by originating from the endosomal system. Exosomes are 40–150 nm, endosome-derived small EVs (sEVs) that are released by cells into the extracellular environment. This review focuses on the biological properties of immune cell-derived sEVs, including composition and cellular targeting and mechanisms by which these immune cell-derived sEVs influence tumor immunity either by suppressing or promoting tumor growth, are discussed. The final section of this review discusses how the biological properties of immune cell-derived sEVs can be manipulated to improve their immunogenicity.
Collapse
Affiliation(s)
- Chi Li
- Experimental Therapeutics Group, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Howard Donninger
- Experimental Therapeutics Group, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States
| | - John Eaton
- Department of Medicine, University of Louisville, Louisville, KY, United States.,Immuno-Oncology Group, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Kavitha Yaddanapudi
- Immuno-Oncology Group, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States.,Division of Immunotherapy, Department of Surgery, University of Louisville, Louisville, KY, United States.,Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States
| |
Collapse
|
15
|
Zhang C, Li C, Shang X, Lin J, Wang H. Surgery as a Potential Treatment Option for Patients With Stage III Small-Cell Lung Cancer: A Propensity Score Matching Analysis. Front Oncol 2019; 9:1339. [PMID: 31850223 PMCID: PMC6901619 DOI: 10.3389/fonc.2019.01339] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 11/15/2019] [Indexed: 01/10/2023] Open
Abstract
Surgery is commonly recommended for patients with stage I small-cell lung cancer (SCLC), whereas chemotherapy and radiotherapy are considered the standard treatment for patients with stage III SCLC. However, recent studies have suggested that a small proportion of patients with SCLC at an advanced stage may benefit from surgical resection. Therefore, in this study, we investigated the effectiveness of surgery in patients with stage III SCLC. Patients were selected from the Surveillance, Epidemiology, and End Results (SEER) database between 2004 and 2013. Propensity score matching (PSM) was used to eliminate any clinical bias. The overall survival (OS) was determined using the Kaplan–Meier method and compared using the log-rank test. The Cox proportional-hazards model was used to identify the effect of surgery on the OS. Of 9606 patients with stage III SCLC, 234 underwent surgery. Compared with the non-surgical group, a higher proportion of patients undergoing surgery had T1 and N0-N1 disease (risen by 10.7% for T1; 12.6% for N0-N1) and a lower proportion had T4 and N3 disease (decreased by 14.3% for T4; 12.5% for N3). The Kaplan-Meier analysis showed that patients who underwent surgery had a better OS before and after PSM. The multivariate analysis showed that surgery was beneficial for patients with stage III SCLC (HR: 0.651, 95% CI 0.524–0.808, P < 0.001). In conclusion, surgical resection might be associated with improved OS for patients with stage III SCLC and may be considered for the treatment of these patients. Further prospective studies are required to confirm these findings.
Collapse
Affiliation(s)
- Chenyue Zhang
- Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Cheng Li
- NHC Key Laboratory of Health Economics and Policy Research, School of Health Care Management, Shandong University, Jinan, China.,Department of Dean's Office, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaoling Shang
- Department of Clinical Laboratory, Qilu Medical College, Shandong University, Jinan, China
| | - Jiamao Lin
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
16
|
Zhang Z, Sang W, Xie L, Dai Y. Metal-organic frameworks for multimodal bioimaging and synergistic cancer chemotherapy. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.213022] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
17
|
Polansky H, Schwab H. How latent viruses cause breast cancer: An explanation based on the microcompetition model. Bosn J Basic Med Sci 2019; 19:221-226. [PMID: 30579323 DOI: 10.17305/bjbms.2018.3950] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/11/2018] [Indexed: 12/23/2022] Open
Abstract
Most breast cancer cases show a decrease in the concentration of the breast cancer type 1 susceptibility protein (BRCA1). However, only a small portion of these cases have a mutated BRCA1 gene. Although many attempts have been made to identify the reason for the decrease in BRCA1 concentration in sporadic, non-heritable breast cancer cases, the cause is still unknown. In this review, we use the Microcompetition Model to explain how certain latent viruses, which are frequently detected in breast cancer tumors, can decrease the expression of the BRCA1 gene and cause the development of breast tumors.
Collapse
Affiliation(s)
- Hanan Polansky
- The Center for the Biology of Chronic Disease (CBCD), New York, NY, USA.
| | | |
Collapse
|
18
|
Yang X, Shi Y, Li M, Lu T, Xi J, Lin Z, Jiang W, Guo W, Zhan C, Wang Q. Identification and validation of an immune cell infiltrating score predicting survival in patients with lung adenocarcinoma. J Transl Med 2019; 17:217. [PMID: 31286969 PMCID: PMC6615164 DOI: 10.1186/s12967-019-1964-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 06/25/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Immune infiltration may predict survival and have clinical significance in lung cancer. However, immune signatures derived from immune profiling based on bulk tumor transcriptomes have not been systematically established in lung adenocarcinoma. We aimed to construct an immune cell infiltrating score, using a new algorithm for evaluating immune infiltration, to improve the prognostic model of lung adenocarcinoma. METHODS Public datasets of lung adenocarcinoma from the Gene Expression Omnibus and The Cancer Genome Atlas were adopted as the training and validation cohorts. Fractions of different immune cell subtypes in each sample were estimated using the CIBERSORT algorithm. The immune infiltrating score was further developed by a least absolute shrinkage and selection operator regression model. The prognostic value and clinical relationship of the model was then further explored. RESULTS An immune infiltrating score model was established on the basis of the immune cells in the training cohort. A high score was associated with significantly worse survival in patients with lung adenocarcinoma (P < 0.001). The prognostic value of the score was confirmed in the validation cohort. The immune infiltrating score could improve the accuracy of predictions of survival when combined with the staging system. Furthermore, the score was potentially associated with patient smoking status and histologic subtype of lung adenocarcinoma. Its possible association with the efficacy of adjuvant chemotherapy was not statistically significant. CONCLUSION The immune cell infiltrating score has prognostic significance in predicting overall survival in patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Xiaodong Yang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Yu Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Ming Li
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Tao Lu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Junjie Xi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Zongwu Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Wei Jiang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Weigang Guo
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China.
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| |
Collapse
|
19
|
Liu H, Zou M, Li P, Wang H, Lin X, Ye J. Oxymatrine‑mediated maturation of dendritic cells leads to activation of FOXP3+/CD4+ Treg cells and reversal of cisplatin‑resistance in lung cancer cells. Mol Med Rep 2019; 19:4081-4090. [PMID: 30896871 PMCID: PMC6471056 DOI: 10.3892/mmr.2019.10064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
The dendritic cell (DC)‑regulatory T (Treg) system serves a leading role in the immunosuppression of the tumor microenvironment, which is not conducive to radiotherapy and chemotherapy treatment for lung cancer. The present study aimed to investigate the effect of oxymatrine (OMT) on the DC‑Treg system in the tumor microenvironment in vitro and to examine its mechanism. The expressions of CD83 antigen, T‑lymphocyte activation antigen CD86, CD11 antigen‑like family member C and major histocompatibility complex II in DCs were increased upon treatment with 1 mg/ml OMT, as detected by flow cytometry. Following pretreatment with OMT, the DCs mediated the forkhead box protein P3 overexpression in primitive cluster of differentiation 4+ T cells at the protein and mRNA expression levels. The expression levels of anti‑inflammatory factors, including interleukin (IL)‑10, tumor growth factor‑β, IL‑35, and pro‑inflammatory cytokines, including interferon‑γ, IL‑12 and IL‑2, in the co‑culture supernatant were increased as measured by ELISA. When DCs and DC‑Tregs were co‑cultured with cisplatin‑resistant A549 cells, the proportion of apoptosis in the co‑culture groups was increased under treatment with cisplatin, which was detected by Annexin V/propidium Iodide staining and western blotting. The present results suggested that OMT may promote the maturation of DCs, mediate the differentiation of T cells into Treg cells, and reverse the resistance of tumor cells to cisplatin in vitro. It was suggested that OMT is an important adjunct to chemotherapy through the regulation of antitumor responses.
Collapse
Affiliation(s)
- Hui Liu
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| | - Manman Zou
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| | - Pei Li
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| | - Haifeng Wang
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| | - Xijun Lin
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| | - Jin Ye
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 501630, P.R. China
| |
Collapse
|
20
|
Belderbos RA, Aerts JGJV, Vroman H. Enhancing Dendritic Cell Therapy in Solid Tumors with Immunomodulating Conventional Treatment. MOLECULAR THERAPY-ONCOLYTICS 2019; 13:67-81. [PMID: 31020037 PMCID: PMC6475716 DOI: 10.1016/j.omto.2019.03.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Dendritic cells (DCs) are the most potent antigen-presenting cells and are the key initiator of tumor-specific immune responses. These characteristics are exploited by DC therapy, where DCs are ex vivo loaded with tumor-associated antigens (TAAs) and used to induce tumor-specific immune responses. Unfortunately, clinical responses remain limited to a proportion of the patients. Tumor characteristics and the immunosuppressive tumor microenvironment (TME) of the tumor are likely hampering efficacy of DC therapy. Therefore, reducing the immunosuppressive TME by combining DC therapy with other treatments could be a promising strategy. Initially, conventional cancer therapies, such as chemotherapy and radiotherapy, were thought to specifically target cancerous cells. Recent insights indicate that these therapies additionally augment tumor immunity by targeting immunosuppressive cell subsets in the TME, inducing immunogenic cell death (ICD), or blocking inhibitory molecules. Therefore, combining DC therapy with registered therapies such as chemotherapy, radiotherapy, or checkpoint inhibitors could be a promising treatment strategy to improve the efficacy of DC therapy. In this review, we evaluate various clinical applicable combination strategies to improve the efficacy of DC therapy.
Collapse
Affiliation(s)
- Robert A Belderbos
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, the Netherlands.,Erasmus MC Cancer Institute, Erasmus MC Rotterdam, the Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, the Netherlands.,Erasmus MC Cancer Institute, Erasmus MC Rotterdam, the Netherlands
| | - Heleen Vroman
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, the Netherlands.,Erasmus MC Cancer Institute, Erasmus MC Rotterdam, the Netherlands
| |
Collapse
|
21
|
Polansky H, Schwab H. Latent viruses can cause disease by disrupting the competition for the limiting factor p300/CBP. Cell Mol Biol Lett 2018; 23:56. [PMID: 30505323 PMCID: PMC6260892 DOI: 10.1186/s11658-018-0121-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/07/2018] [Indexed: 12/11/2022] Open
Abstract
CBP and p300 are histone acetyltransferase coactivators that control the transcription of numerous genes in humans, viruses, and other organisms. Although two separate genes encode CBP and p300, they share a 61% sequence identity, and they are often mentioned together as p300/CBP. Zhou et al. showed that under hypoxic conditions, HIF1α and the tumor suppressor p53 compete for binding to the limiting p300/CBP coactivator. Jethanandani & Kramer showed that δEF1 and MYOD genes compete for the limited amount of p300/CBP in the cell. Bhattacharyya et al. showed that the limiting availability of p300/CBP in the cell serves as a checkpoint for HIF1α activity. Here, we use the microcompetition model to explain how latent viruses with a specific viral cis-regulatory element in their promoter/enhancer can disrupt this competition, causing diseases such as cancer, diabetes, atherosclerosis, and obesity.
Collapse
Affiliation(s)
- Hanan Polansky
- The Center for the Biology of Chronic Disease (CBCD), 616 Corporate Way, Suite 2-3665, Valley Cottage, New York City, NY 10989 USA
| | - Hava Schwab
- The Center for the Biology of Chronic Disease (CBCD), 616 Corporate Way, Suite 2-3665, Valley Cottage, New York City, NY 10989 USA
| |
Collapse
|
22
|
De Mattia E, Dreussi E, Montico M, Gagno S, Zanusso C, Quartuccio L, De Vita S, Guardascione M, Buonadonna A, D'Andrea M, Pella N, Favaretto A, Mini E, Nobili S, Romanato L, Cecchin E, Toffoli G. A Clinical-Genetic Score to Identify Surgically Resected Colorectal Cancer Patients Benefiting From an Adjuvant Fluoropyrimidine-Based Therapy. Front Pharmacol 2018; 9:1101. [PMID: 30337874 PMCID: PMC6180157 DOI: 10.3389/fphar.2018.01101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/10/2018] [Indexed: 12/20/2022] Open
Abstract
There are clinical challenges related to adjuvant treatment in colorectal cancer (CRC) and novel molecular markers are needed for better risk stratification of patients. Our aim was to integrate our previously reported clinical-genetic prognostic score with new immunogenetic markers of 5-year disease-free survival (DFS) to evaluate the recurrence risk stratification before fluoropyrimidine (FL)-based adjuvant therapy. The study population included a total of 270 stage II-III CRC patients treated with adjuvant FL with (FL + OXA, n = 119) or without oxaliplatin (FL, n = 151). Patients were genotyped for a panel of 192 tagging polymorphisms in 34 immune-related genes. The IFNG-rs1861494 polymorphism was associated with worse DFS in the FL + OXA (HR = 2.14, 95%CI 1.13–4.08; P = 0.020, q-value = 0.249) and FL (HR = 1.97, 95%CI 1.00–3.86; P = 0.049) cohorts, according to a dominant model. The integration of IFNG-rs1861494 in our previous clinical genetic multiparametric score of DFS improved the patients’ risk stratification (Log-rank P = 0.0026 in the pooled population). These findings could improve the discrimination of patients who would benefit from adjuvant treatment. In addition, the results may help better elucidate the interplay between the immune system and chemotherapeutics and help determine the efficacy of anti-tumor strategies.
Collapse
Affiliation(s)
- Elena De Mattia
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Eva Dreussi
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Marcella Montico
- Scientific Directorate, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Sara Gagno
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Chiara Zanusso
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Luca Quartuccio
- Department of Medical Area (DAME), Rheumatology Clinic, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Salvatore De Vita
- Department of Medical Area (DAME), Rheumatology Clinic, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Michela Guardascione
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Angela Buonadonna
- Medical Oncology Unit B, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Mario D'Andrea
- Medical Oncology Unit, "San Filippo Neri Hospital", Rome, Italy
| | | | | | - Enrico Mini
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Stefania Nobili
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Loredana Romanato
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| |
Collapse
|
23
|
Cecchin E, De Mattia E, Ecca F, Toffoli G. Host genetic profiling to increase drug safety in colorectal cancer from discovery to implementation. Drug Resist Updat 2018; 39:18-40. [PMID: 30075835 DOI: 10.1016/j.drup.2018.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/11/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023]
Abstract
Adverse events affect the pharmacological treatment of approximately 90% of colorectal cancer (CRC) patients at any stage of the disease. Chemotherapy including fluoropyrimidines, irinotecan, and oxaliplatin is the cornerstone of the pharmacological treatment of CRC. The introduction of novel targeted agents, as anti-EGFR (i.e. cetuximab, panitumumab) and antiangiogenic (i.e. bevacizumab, ziv-aflibercept, regorafenib, and ramucirumab) molecules, into the oncologist's toolbox has led to significant improvements in the life expectancy of advanced CRC patients, but with a substantial increase in toxicity burden. In this respect, pharmacogenomics has largely been applied to the personalization of CRC chemotherapy, focusing mainly on the study of inhered polymorphisms in genes encoding phase I and II enzymes, ATP-binding cassette (ABC)/solute carrier (SLC) membrane transporters, proteins involved in DNA repair, folate pathway and immune response. These research efforts have led to the identification of some validated genetic markers of chemotherapy toxicity, for fluoropyrimidines and irinotecan. No validated genetic determinants of oxaliplatin-specific toxicity, as peripheral neuropathy, has thus far been established. The contribution of host genetic markers in predicting the toxicity associated with novel targeted agents' administration is still controversial due to the heterogeneity of published data. Pharmacogenomics guidelines have been published by some international scientific consortia such as the Clinical Pharmacogenomics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG) strongly suggesting a pre-treatment dose adjustment of irinotecan based on UGT1A1*28 genotype and of fluoropyrimidines based on some DPYD genetic variants, to increase treatment safety. However, these recommendations are still poorly applied at the patient's bedside. Several ongoing projects in the U.S. and Europe are currently evaluating how pharmacogenomics can be implemented successfully in daily clinical practice. The majority of drug-related adverse events are still unexplained, and a great deal of ongoing research is aimed at improving knowledge of the role of pharmacogenomics in increasing treatment safety. In this review, the issue of pre-treatment identification of CRC patients at risk of toxicity via the analysis of patients' genetic profiles is addressed. Available pharmacogenomics guidelines with ongoing efforts to implement them in clinical practice and new exploratory markers for clinical validation are described.
Collapse
Affiliation(s)
- Erika Cecchin
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Fabrizio Ecca
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy.
| |
Collapse
|
24
|
Zapater-Moros A, Gámez-Pozo A, Prado-Vázquez G, Trilla-Fuertes L, Arevalillo JM, Díaz-Almirón M, Navarro H, Maín P, Feliú J, Zamora P, Espinosa E, Fresno Vara JÁ. Probabilistic graphical models relate immune status with response to neoadjuvant chemotherapy in breast cancer. Oncotarget 2018; 9:27586-27594. [PMID: 29963222 PMCID: PMC6021258 DOI: 10.18632/oncotarget.25496] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 05/08/2018] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is the most frequent tumor in women and its incidence is increasing. Neoadjuvant chemotherapy has become standard of care as a complement to surgery in locally advanced or poor-prognosis early stage disease. The achievement of a complete response to neoadjuvant chemotherapy correlates with prognosis but it is not possible to predict who will obtain an excellent response. The molecular analysis of the tumor offers a unique opportunity to unveil predictive factors. In this work, gene expression profiling in 279 tumor samples from patients receiving neoadjuvant chemotherapy was performed and probabilistic graphical models were used. This approach enables addressing biological and clinical questions from a Systems Biology perspective, allowing to deal with large gene expression data and their interactions. Tumors presenting complete response to neoadjuvant chemotherapy had a higher activity of immune related functions compared to resistant tumors. Similarly, samples from complete responders presented higher expression of lymphocyte cell lineage markers, immune-activating and immune-suppressive markers, which may correlate with tumor infiltration by lymphocytes (TILs). These results suggest that the patient's immune system plays a key role in tumor response to neoadjuvant treatment. However, future studies with larger cohorts are necessary to validate these hypotheses.
Collapse
Affiliation(s)
- Andrea Zapater-Moros
- Molecular Oncology & Pathology Laboratory, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Madrid, Spain
| | - Angelo Gámez-Pozo
- Molecular Oncology & Pathology Laboratory, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Madrid, Spain
- Biomedica Molecular Medicine SL, Madrid, Spain
| | - Guillermo Prado-Vázquez
- Molecular Oncology & Pathology Laboratory, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Madrid, Spain
| | | | - Jorge M. Arevalillo
- Operational Research and Numerical Analysis, National Distance Education University, Madrid, Spain
| | | | - Hilario Navarro
- Operational Research and Numerical Analysis, National Distance Education University, Madrid, Spain
| | - Paloma Maín
- Department of Statistics and Operations Research, Faculty of Mathematics, Complutense University of Madrid, Madrid, Spain
| | - Jaime Feliú
- Medical Oncology Service, La Paz University Hospital-IdiPAZ, Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Pilar Zamora
- Medical Oncology Service, La Paz University Hospital-IdiPAZ, Madrid, Spain
| | - Enrique Espinosa
- Medical Oncology Service, La Paz University Hospital-IdiPAZ, Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Juan Ángel Fresno Vara
- Molecular Oncology & Pathology Laboratory, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Madrid, Spain
- Biomedica Molecular Medicine SL, Madrid, Spain
- CIBERONC, Madrid, Spain
| |
Collapse
|
25
|
Mitchell LA, Lopez Espinoza F, Mendoza D, Kato Y, Inagaki A, Hiraoka K, Kasahara N, Gruber HE, Jolly DJ, Robbins JM. Toca 511 gene transfer and treatment with the prodrug, 5-fluorocytosine, promotes durable antitumor immunity in a mouse glioma model. Neuro Oncol 2018; 19:930-939. [PMID: 28387849 PMCID: PMC5570153 DOI: 10.1093/neuonc/nox037] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background. Toca 511 (vocimagene amiretrorepvec) is a retroviral replicating vector encoding an optimized yeast cytosine deaminase (CD). Tumor-selective expression of CD converts the prodrug, 5-fluorocytosine (5-FC), into the active chemotherapeutic, 5-fluorouracil (5-FU). This therapeutic approach is being tested in a randomized phase II/III trial in recurrent glioblastoma and anaplastic astrocytoma (NCT0241416). The aim of this study was to identify the immune cell subsets contributing to antitumor immune responses following treatment with 5-FC in Toca 511–expressing gliomas in a syngeneic mouse model. Methods. Flow cytometry was utilized to monitor and characterize the immune cell infiltrate in subcutaneous Tu-2449 gliomas in B6C3F1 mice treated with Toca 511 and 5-FC. Results. Tumor-bearing animals treated with Toca 511 and 5-FC display alterations in immune cell populations within the tumor that result in antitumor immune protection. Attenuated immune subsets were exclusive to immunosuppressive cells of myeloid origin. Depletion of immunosuppressive cells temporally preceded a second event which included expansion of T cells which were polarized away from Th2 and Th17 in the CD4+ T cell compartment with concomitant expansion of interferon gamma–expressing CD8+ T cells. Immune alterations correlated with clearance of Tu-2449 subcutaneous tumors and T cell–dependent protection from future tumor challenge. Conclusions. Treatment with Toca 511 and 5-FC has a concentrated effect at the site of the tumor which causes direct tumor cell death and alterations in immune cell infiltrate, resulting in a tumor microenvironment that is more permissive to establishment of a T cell mediated antitumor immune response.
Collapse
Affiliation(s)
- Leah A Mitchell
- Tocagen Inc., San Diego, California; DNAtrix Inc., Houston, Texas; University of California Los Angeles, Los Angeles, California; Department of Gastroenterological Surgery, Hokkaido University, Sapporo, Japan; Department of Cell Biology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Fernando Lopez Espinoza
- Tocagen Inc., San Diego, California; DNAtrix Inc., Houston, Texas; University of California Los Angeles, Los Angeles, California; Department of Gastroenterological Surgery, Hokkaido University, Sapporo, Japan; Department of Cell Biology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Daniel Mendoza
- Tocagen Inc., San Diego, California; DNAtrix Inc., Houston, Texas; University of California Los Angeles, Los Angeles, California; Department of Gastroenterological Surgery, Hokkaido University, Sapporo, Japan; Department of Cell Biology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Yuki Kato
- Tocagen Inc., San Diego, California; DNAtrix Inc., Houston, Texas; University of California Los Angeles, Los Angeles, California; Department of Gastroenterological Surgery, Hokkaido University, Sapporo, Japan; Department of Cell Biology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Akihito Inagaki
- Tocagen Inc., San Diego, California; DNAtrix Inc., Houston, Texas; University of California Los Angeles, Los Angeles, California; Department of Gastroenterological Surgery, Hokkaido University, Sapporo, Japan; Department of Cell Biology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Kei Hiraoka
- Tocagen Inc., San Diego, California; DNAtrix Inc., Houston, Texas; University of California Los Angeles, Los Angeles, California; Department of Gastroenterological Surgery, Hokkaido University, Sapporo, Japan; Department of Cell Biology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Noriyuki Kasahara
- Tocagen Inc., San Diego, California; DNAtrix Inc., Houston, Texas; University of California Los Angeles, Los Angeles, California; Department of Gastroenterological Surgery, Hokkaido University, Sapporo, Japan; Department of Cell Biology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Harry E Gruber
- Tocagen Inc., San Diego, California; DNAtrix Inc., Houston, Texas; University of California Los Angeles, Los Angeles, California; Department of Gastroenterological Surgery, Hokkaido University, Sapporo, Japan; Department of Cell Biology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Douglas J Jolly
- Tocagen Inc., San Diego, California; DNAtrix Inc., Houston, Texas; University of California Los Angeles, Los Angeles, California; Department of Gastroenterological Surgery, Hokkaido University, Sapporo, Japan; Department of Cell Biology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Joan M Robbins
- Tocagen Inc., San Diego, California; DNAtrix Inc., Houston, Texas; University of California Los Angeles, Los Angeles, California; Department of Gastroenterological Surgery, Hokkaido University, Sapporo, Japan; Department of Cell Biology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
26
|
Rangan L, Galaine J, Boidot R, Hamieh M, Dosset M, Francoual J, Beziaud L, Pallandre JR, Lauret Marie Joseph E, Asgarova A, Borg C, Al Saati T, Godet Y, Latouche JB, Valmary-Degano S, Adotévi O. Identification of a novel PD-L1 positive solid tumor transplantable in HLA-A*0201/DRB1*0101 transgenic mice. Oncotarget 2018; 8:48959-48971. [PMID: 28430664 PMCID: PMC5564740 DOI: 10.18632/oncotarget.16900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 03/22/2017] [Indexed: 12/28/2022] Open
Abstract
HLA-A*0201/DRB1*0101 transgenic mice (A2/DR1 mice) have been developed to study the
immunogenicity of tumor antigen-derived T cell epitopes. To extend the use and
application of this mouse model in the field of antitumor immunotherapy, we described
a tumor cell line generated from a naturally occurring tumor in A2/DR1 mouse named
SARC-L1. Histological and genes signature analysis supported the sarcoma origin of
this cell line. While SARC-L1 tumor cells lack HLA-DRB1*0101 expression, a very low
expression of HLA-A*0201 molecules was found on these cells. Furthermore they also
weakly but constitutively expressed the programmed death-ligand 1 (PD-L1).
Interestingly both HLA-A*0201 and PD-L1 expressions can be increased on SARC-L1 after
IFN-γ exposure in vitro. We also obtained two genetically
modified cell lines highly expressing either HLA-A*0201 or both HLA-A*0201/
HLA-DRB1*0101 molecules referred as SARC-A2 and SARC-A2DR1 respectively. All the
SARC-L1-derived cell lines induced aggressive subcutaneous tumors in A2DR1 mice
in vivo. The analysis of SARC-L1 tumor microenvironment revealed
a strong infiltration by T cells expressing inhibitory receptors such as PD-1 and
TIM-3. Finally, we found that SARC-L1 is sensitive to several drugs commonly used to
treat sarcoma and also susceptible to anti-PD-L1 monoclonal antibody therapy
in vivo. Collectively, we described a novel syngeneic tumor model
A2/DR1 mice that could be used as preclinical tool for the evaluation of antitumor
immunotherapies.
Collapse
Affiliation(s)
- Laurie Rangan
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-gGreffon-Tumeur, Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,LabEx LipSTIC, F-25000 Besançon, France
| | - Jeanne Galaine
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-gGreffon-Tumeur, Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,LabEx LipSTIC, F-25000 Besançon, France
| | - Romain Boidot
- Platform for Transfer to Cancer Biology, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Mohamad Hamieh
- University Hospital of Rouen, INSERM UMR1245, Institute for Research and Innovation in Biomedicine, 76183 Rouen, France
| | - Magalie Dosset
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-gGreffon-Tumeur, Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,LabEx LipSTIC, F-25000 Besançon, France
| | - Julie Francoual
- University Hospital of Rouen, INSERM UMR1245, Institute for Research and Innovation in Biomedicine, 76183 Rouen, France
| | - Laurent Beziaud
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-gGreffon-Tumeur, Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,LabEx LipSTIC, F-25000 Besançon, France
| | - Jean-René Pallandre
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-gGreffon-Tumeur, Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - Elodie Lauret Marie Joseph
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-gGreffon-Tumeur, Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,LabEx LipSTIC, F-25000 Besançon, France
| | - Afag Asgarova
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-gGreffon-Tumeur, Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,LabEx LipSTIC, F-25000 Besançon, France
| | - Christophe Borg
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-gGreffon-Tumeur, Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,LabEx LipSTIC, F-25000 Besançon, France.,Department of Medical Oncology, University Hospital of Besançon, 25000 Besançon, France
| | - Talal Al Saati
- INSERM/UPS, US006/CREFRE, Department of Histopathology, University Hospital of Purpan, 31000 Toulouse, France
| | - Yann Godet
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-gGreffon-Tumeur, Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,LabEx LipSTIC, F-25000 Besançon, France
| | - Jean Baptiste Latouche
- Department of Genetics, University Hospital of Rouen, Normandy Centre for Genomic and Personalized Medicine, 76183 Rouen, France
| | | | - Olivier Adotévi
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-gGreffon-Tumeur, Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,LabEx LipSTIC, F-25000 Besançon, France.,Department of Medical Oncology, University Hospital of Besançon, 25000 Besançon, France
| |
Collapse
|
27
|
Van Acker HH, Anguille S, De Reu H, Berneman ZN, Smits EL, Van Tendeloo VF. Interleukin-15-Cultured Dendritic Cells Enhance Anti-Tumor Gamma Delta T Cell Functions through IL-15 Secretion. Front Immunol 2018; 9:658. [PMID: 29692776 PMCID: PMC5902500 DOI: 10.3389/fimmu.2018.00658] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/16/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cell (DC) vaccination can be an effective post-remission therapy for acute myeloid leukemia (AML). Yet, current DC vaccines do not encompass the ideal stimulatory triggers for innate gamma delta (γδ) T cell anti-tumor activity. Promoting type 1 cytotoxic γδ T cells in patients with AML is, however, most interesting, considering these unconventional T cells are primed for rapid function and exert meaningful control over AML. In this work, we demonstrate that interleukin (IL)-15 DCs have the capacity to enhance the anti-tumoral functions of γδ T cells. IL-15 DCs of healthy donors and of AML patients in remission induce the upregulation of cytotoxicity-associated and co-stimulatory molecules on the γδ T cell surface, but not of co-inhibitory molecules, incite γδ T cell proliferation and stimulate their interferon-γ production in the presence of blood cancer cells and phosphoantigens. Moreover, the innate cytotoxic capacity of γδ T cells is significantly enhanced upon interaction with IL-15 DCs, both towards leukemic cell lines and allogeneic primary AML blasts. Finally, we address soluble IL-15 secreted by IL-15 DCs as the main mechanism behind the IL-15 DC-mediated γδ T cell activation. These results indicate that the application of IL-15-secreting DC subsets could render DC-based anti-cancer vaccines more effective through, among others, the involvement of γδ T cells in the anti-leukemic immune response.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Sébastien Anguille
- Division of Hematology, Antwerp University Hospital, Edegem, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Division of Hematology, Antwerp University Hospital, Edegem, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium.,Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
28
|
Menter T, Tzankov A. Mechanisms of Immune Evasion and Immune Modulation by Lymphoma Cells. Front Oncol 2018; 8:54. [PMID: 29564225 PMCID: PMC5845888 DOI: 10.3389/fonc.2018.00054] [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: 12/22/2017] [Accepted: 02/20/2018] [Indexed: 02/06/2023] Open
Abstract
Purpose Targeting cancer cells by modulating the immune system has become an important new therapeutic option in many different malignancies. Inhibition of CTLA4/B7 and PD1/PDL1 signaling is now also being investigated and already successfully applied to various hematologic malignancies. Methods A literature review of PubMed and results of our own studies were compiled in order to give a comprehensive overview on this topic. Results We elucidate the pathophysiological role of immunosuppressive networks in lymphomas, ranging from changes in the cellular microenvironment composition to distinct signaling pathways such as PD1/PDL1 or CTLA4/B7/CD28. The prototypical example of a lymphoma manipulating and thereby silencing the immune system is Hodgkin lymphoma. Also other lymphomas, e.g., primary mediastinal B-cell lymphoma and some Epstein–Barr virus (EBV)-driven malignancies, use analogous survival strategies, while diffuse large B-cell lymphoma of the activated B-cell type, follicular lymphoma and angioimmunoblastic T-cell lymphoma to name a few, exert further immune escape strategies each. These insights have already led to new treatment opportunities and results of the most important clinical trials based on this concept are briefly summarized. Immune checkpoint inhibition might also have severe side effects; the mechanisms of the rather un(der)recognized hematological side effects of this treatment approach are discussed. Conclusion Silencing the host’s immune system is an important feature of various lymphomas. Achieving a better understanding of distinct pathways of interactions between lymphomas and different immunological microenvironment compounds yields substantial potential for new treatment concepts.
Collapse
Affiliation(s)
- Thomas Menter
- Institute of Pathology and Medical Genetics, University Hospital of Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Pathology and Medical Genetics, University Hospital of Basel, Basel, Switzerland
| |
Collapse
|
29
|
Kather JN, Halama N, Jaeger D. Genomics and emerging biomarkers for immunotherapy of colorectal cancer. Semin Cancer Biol 2018; 52:189-197. [PMID: 29501787 DOI: 10.1016/j.semcancer.2018.02.010] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/19/2018] [Accepted: 02/28/2018] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is a common and lethal disease with a high therapeutic need. For most patients with metastatic CRC, chemotherapy is the only viable option. Currently, immunotherapy is restricted to the particular genetic subgroup of mismatch-repair deficient (MMRd)/microsatellite instable (MSI) CRC. Anti-PD1 therapy was recently FDA-approved as a second-line treatment in this subgroup. However, in a metastatic setting, these MMRd/MSI tumors are vastly outnumbered by mismatch-repair proficient (MMRp)/microsatellite stable (MSS) tumors. These MMRp/MSS tumors do not meaningfully respond to any traditional immunotherapy approach including checkpoint blockade, adoptive cell transfer and vaccination. This resistance to immunotherapy is due to a complex tumor microenvironment that counteracts antitumor immunity through a combination of poorly antigenic tumor cells and an immunosuppressive tumor microenvironment. To find ways of overcoming immunotherapy resistance in the majority of CRC patients, it is necessary to analyze the immunological makeup in an in-depth and personalized way and in the context of their tumor genetic makeup. Flexible, biomarker-guided early-phase immunotherapy trials are needed to optimize this workflow. In this review, we detail key mechanisms for immune evasion and emerging immune biomarkers for personalized immunotherapy in CRC. Also, we present a template for biomarker-guided clinical trials that are needed to move new immunotherapy approaches closer to clinical application.
Collapse
Affiliation(s)
- Jakob Nikolas Kather
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Niels Halama
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Dirk Jaeger
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
30
|
PD-L1 Expression in TNBC: A Predictive Biomarker of Response to Neoadjuvant Chemotherapy? BIOMED RESEARCH INTERNATIONAL 2017; 2017:1750925. [PMID: 29387716 PMCID: PMC5745649 DOI: 10.1155/2017/1750925] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 11/19/2017] [Indexed: 11/25/2022]
Abstract
Triple negative breast cancer (TNBC) has an aggressive clinical behaviour, with a poorer prognosis compared to other subtypes. Recently, tumor-infiltrating lymphocytes (TILs) have been proposed as a predictive biomarker for a better clinical outcome and pathological response (pR) after neoadjuvant chemotherapy (NACT) in TNBC. These data confirm the role of the immune system in the neoplastic progression and in the response to therapy. We performed a retrospective analysis of 54 pre-NACT biopsies of TNBC and compared both the percentage of stromal TILs and the degree of PD-L1 expression with the extent of pR to standard NACT. A pathological complete response (pCR) was achieved in 35% of cases. Univariate analysis showed (i) a significant association between PD-L1 expression in ≥25% of neoplastic cells and the achievement of a pCR (p = 0.024); (ii) a significantly higher frequency of pCR in cases showing ≥50% stromal TILs (p < 0.001). However in the multivariate analysis only PD-L1 expression on tumor cells remained significantly associated with pCR (OR = 1,13; 95% CI 1,01–1,27), suggesting that the expression of this biomarker could be associated with a subpopulation of TNBC more likely to respond to chemotherapy. These data need to be confirmed by larger studies.
Collapse
|
31
|
Jacobs J, Deschoolmeester V, Rolfo C, Zwaenepoel K, Van den Bossche J, Deben C, Silence K, de Haard H, Hermans C, Rottey S, Vangestel C, Lardon F, Smits E, Pauwels P. Preclinical data on the combination of cisplatin and anti-CD70 therapy in non-small cell lung cancer as an excellent match in the era of combination therapy. Oncotarget 2017; 8:74058-74067. [PMID: 29088768 PMCID: PMC5650323 DOI: 10.18632/oncotarget.18202] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/12/2017] [Indexed: 12/04/2022] Open
Abstract
In contrast to the negligible expression of the immunomodulating protein CD70 in normal tissue, we have demonstrated constitutive overexpression of CD70 on tumor cells in a subset of primary non-small cell lung cancer (NSCLC) biopsies. This can be exploited by CD70-targeting antibody-dependent cellular cytotoxicity (ADCC)-inducing antibodies. Early clinical trials of these antibodies have already shown promising results in CD70-positive malignancies. In this study, we explored the potential of cisplatin to induce CD70 expression in NSCLC. Using real-time measurement tools, we also assessed the efficacy of a combination regimen with cisplatin and anti-CD70 therapy under normoxia and hypoxia. We identified an induction of CD70 expression on lung cancer cells upon low doses of cisplatin, independent of oxygen levels. More importantly, the use of cisplatin resulted in an enhanced ADCC-effect of anti-CD70 therapy. As such, this combination regimen led to a significant decrease in lung cancer cell survival cell survival, broadening the applicability the applicability of CD70-targeting therapy. This is the first study that proves the potential of a combination therapy with cisplatin and CD70-targeting drugs in NSCLC. Based on our data, we postulate that this combination strategy is an interesting approach to increase tumor-specific cytotoxicity and reduce drug-related side effects.
Collapse
Affiliation(s)
- Julie Jacobs
- Center for Oncological Research, University of Antwerp, Antwerp, 2610 Wilrijk, Belgium.,Department of Pathology, Antwerp University Hospital, Antwerp, 2650 Edegem, Belgium
| | - Vanessa Deschoolmeester
- Center for Oncological Research, University of Antwerp, Antwerp, 2610 Wilrijk, Belgium.,Department of Pathology, Antwerp University Hospital, Antwerp, 2650 Edegem, Belgium
| | - Christian Rolfo
- Department of Oncology, Antwerp University Hospital, Antwerp, 2650, Edegem, Belgium.,Phase 1-Early Clinical Trials Unit, Antwerp University Hospital, Antwerp, 2650 Edegem, Belgium
| | - Karen Zwaenepoel
- Center for Oncological Research, University of Antwerp, Antwerp, 2610 Wilrijk, Belgium.,Department of Pathology, Antwerp University Hospital, Antwerp, 2650 Edegem, Belgium
| | | | - Christophe Deben
- Center for Oncological Research, University of Antwerp, Antwerp, 2610 Wilrijk, Belgium.,Department of Pathology, Antwerp University Hospital, Antwerp, 2650 Edegem, Belgium
| | | | | | - Christophe Hermans
- Center for Oncological Research, University of Antwerp, Antwerp, 2610 Wilrijk, Belgium.,Department of Pathology, Antwerp University Hospital, Antwerp, 2650 Edegem, Belgium
| | - Sylvie Rottey
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Christel Vangestel
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, 2610 Wilrijk, Belgium
| | - Filip Lardon
- Center for Oncological Research, University of Antwerp, Antwerp, 2610 Wilrijk, Belgium
| | - Evelien Smits
- Center for Oncological Research, University of Antwerp, Antwerp, 2610 Wilrijk, Belgium.,Laboratory of Experimental Hematology (LEH), Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, 2610 Wilrijk, Belgium
| | - Patrick Pauwels
- Center for Oncological Research, University of Antwerp, Antwerp, 2610 Wilrijk, Belgium.,Department of Pathology, Antwerp University Hospital, Antwerp, 2650 Edegem, Belgium
| |
Collapse
|
32
|
Exosomes miR-126a released from MDSC induced by DOX treatment promotes lung metastasis. Oncogene 2016; 36:639-651. [PMID: 27345402 PMCID: PMC5419051 DOI: 10.1038/onc.2016.229] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 03/14/2016] [Accepted: 05/22/2016] [Indexed: 12/13/2022]
Abstract
Acquired resistance to chemotherapy remains a major stumbling block in cancer treatment. Chronic inflammation plays a crucial role in induction of chemo resistance, and results in part from the induction and expansion of inflammatory cells that include myeloid derived suppressor cells (MDSC) and IL-13+Th2 cells. The mechanisms that lead to induction of activated MDSCs and IL-13+Th2 cells have not yet been identified. Here we demonstrated that doxorubicin treatment of 4T1 breast tumor bearing mice led to the induction of IL-13R+miR-126a+MDSC (DOX-MDSC). DOX-MDSC promote breast tumor lung metastasis through MDSC miR-126a+exosomal mediated induction of IL-13+Th2 cells and tumor angiogenesis. The induction of DOX-MDSC is regulated in a paracrine manner. DOX treatment not only increases IL-33 released from breast tumor cells, which is crucial for the induction of IL-13+Th2 cells, but it also participates in the induction of IL-13 receptors and miR-126a expressed on/in the MDSCs. IL-13 released from IL-13+Th2 cells then promotes the production of DOX-MDSC and MDSC miR-126a+exosomes via MDSC IL-13R. MDSC miR-126a+exosomes further induce IL13+Th2 cells in a positive feed-back loop manner. We also showed that MDSC miR-126a rescues doxorubicin induced MDSC death in a S100A8/A9 dependent manner and promotes tumor angiogenesis. Our findings provide insight into the MDSC exosomal mediated chemo resistance mechanism, which will be useful for the design of inhibitors targeting the blocking of induction of miR-126a+MDSC.
Collapse
|
33
|
Ingold Heppner B, Loibl S, Denkert C. Tumor-Infiltrating Lymphocytes: A Promising Biomarker in Breast Cancer. Breast Care (Basel) 2016; 11:96-100. [PMID: 27239170 DOI: 10.1159/000444357] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There is clear evidence that the immune system plays an essential role in tumor defense. By determining tumor-infiltrating lymphocytes (TILs), the individual immunological response becomes more apparent and measurable. In breast cancer, high levels of TILs are associated with a more favorable clinical course. In this review, we describe how TILs are determined with emphasis on daily routine diagnostics. We further discuss their impact as a prognostic and predictive biomarker in the neoadjuvant and adjuvant therapy setting as well as in residual disease. We also discuss their potential future implications on further stratifying prognostic subgroups of breast cancer, thereby possibly influencing future therapy considerations.
Collapse
Affiliation(s)
| | - Sibylle Loibl
- German Breast Group (GBG) c/o Forschungs GmbH, Neu-Isenburg, Germany; Sana Klinikum Offenbach, Offenbach, Germany
| | - Carsten Denkert
- Institut für Pathologie, Charité Universitätsmedizin, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany
| |
Collapse
|
34
|
|
35
|
Cao G, Wang J, Zheng X, Wei H, Tian Z, Sun R. Tumor Therapeutics Work as Stress Inducers to Enhance Tumor Sensitivity to Natural Killer (NK) Cell Cytolysis by Up-regulating NKp30 Ligand B7-H6. J Biol Chem 2015; 290:29964-73. [PMID: 26472927 DOI: 10.1074/jbc.m115.674010] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Indexed: 11/06/2022] Open
Abstract
Immune cells are believed to participate in initiating anti-tumor effects during regular tumor therapy such as chemotherapy, radiation, hyperthermia, and cytokine injection. One of the mechanisms underlying this process is the expression of so-called stress-inducible immunostimulating ligands. Although the activating receptor NKG2D has been proven to play roles in tumor therapy through targeting its ligands, the role of NKp30, another key activating receptor, is seldom addressed. In this study, we found that the NKp30 ligand B7-H6 was widely expressed in tumor cells and closely correlated to their susceptibility to NK cell lysis. Further studies showed that treatment of tumor cells with almost all standard tumor therapeutics, including chemotherapy (cisplatin, 5-fluorouracil), radiation therapy, non-lethal heat shock, and cytokine therapy (TNF-α), could up-regulate the expression of B7-H6 in tumor cells and enhance tumor sensitivity to NK cell cytolysis. B7-H6 shRNA treatment effectively dampened sensitization of tumor cells to NK-mediated lysis. Our study not only reveals the possibility that tumor therapeutics work as stress inducers to enhance tumor sensitivity to NK cell cytolysis but also suggests that B7-H6 could be a potential target for tumor therapy in the future.
Collapse
Affiliation(s)
- Guoshuai Cao
- From the Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Jian Wang
- From the Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China,
| | - Xiaodong Zheng
- From the Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Haiming Wei
- From the Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China, Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Zhigang Tian
- From the Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China, Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Rui Sun
- From the Institute of Immunology, Key Laboratory of Innate Immunity and Chronic Disease of Chinese Academy of Science, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China, Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China, and
| |
Collapse
|
36
|
Logsdon CD, Arumugam T, Ramachandran V. Animal Models of Gastrointestinal and Liver Diseases. The difficulty of animal modeling of pancreatic cancer for preclinical evaluation of therapeutics. Am J Physiol Gastrointest Liver Physiol 2015; 309:G283-91. [PMID: 26159697 PMCID: PMC4556944 DOI: 10.1152/ajpgi.00169.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/25/2015] [Indexed: 01/31/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is relatively rare but extremely lethal. Standard cytotoxic therapeutics provide little benefit. To date, newer targeted therapeutics have also not been highly successful. Often novel therapeutics that have appeared to perform well in preclinical models have failed in the clinic. Many factors contribute to these failures, but the one most often attributed is the shortcomings of the preclinical models. A plethora of animal models now exist for PDAC, including cell line xenografts, patient-derived xenografts, a wide variety of genetic mouse models, and syngeneic xenografts. These models have generated a tremendous amount of information useful for the understanding of PDAC. Yet none seems to well predict clinical outcomes of new treatments. This review will discuss how genetic instability and cellular heterogeneity make this disease so difficult to model accurately. We will also discuss the strengths and weaknesses of many of the popular models. Ultimately we will argue that there is no perfect model and that the best approach to understanding clinical performance is the use of multiple preclinical models with an understanding of their salient features.
Collapse
Affiliation(s)
- Craig D. Logsdon
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and ,2Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Thiruvengadam Arumugam
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and
| | - Vijaya Ramachandran
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and
| |
Collapse
|
37
|
Dushyanthen S, Beavis PA, Savas P, Teo ZL, Zhou C, Mansour M, Darcy PK, Loi S. Relevance of tumor-infiltrating lymphocytes in breast cancer. BMC Med 2015; 13:202. [PMID: 26300242 PMCID: PMC4547422 DOI: 10.1186/s12916-015-0431-3] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/22/2015] [Indexed: 12/13/2022] Open
Abstract
While breast cancer has not been considered a cancer amenable to immunotherapeutic approaches, recent studies have demonstrated evidence of significant immune cell infiltration via tumor-infiltrating lymphocytes in a subset of patient tumors. In this review we present the current evidence highlighting the clinical relevance and utility of tumor-infiltrating lymphocytes in breast cancer. Retrospective and prospective studies have shown that the presence of tumor-infiltrating lymphocytes is a prognostic marker for higher responses to neoadjuvant chemotherapy and better survival, particularly in triple negative and HER2-positive early breast cancer. Further work is required to determine the immune subsets important in this response and to discover ways of encouraging immune infiltrate in tumor-infiltrating lymphocytes-negative patients.
Collapse
Affiliation(s)
- Sathana Dushyanthen
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Paul A Beavis
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Peter Savas
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Zhi Ling Teo
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Chenhao Zhou
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Mariam Mansour
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Phillip K Darcy
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Sherene Loi
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia. .,Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia.
| |
Collapse
|
38
|
Soliman HH, Jackson E, Neuger T, Dees EC, Harvey RD, Han H, Ismail-Khan R, Minton S, Vahanian NN, Link C, Sullivan DM, Antonia S. A first in man phase I trial of the oral immunomodulator, indoximod, combined with docetaxel in patients with metastatic solid tumors. Oncotarget 2015; 5:8136-46. [PMID: 25327557 PMCID: PMC4226672 DOI: 10.18632/oncotarget.2357] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Indoleamine 2,3-dioxygenase (IDO) is an enzyme that tumors use to create a state of immunosuppression. Indoximod is an IDO pathway inhibitor. Preclinical studies demonstrated that indoximod combined with chemotherapy was synergistic in a mouse model of breast cancer. A phase I 3+3 trial was designed to study the combination of docetaxel and indoximod. METHODS Docetaxel was administered at 60 mg/m2 intravenously every 3 weeks dose levels 1-4 and 75 mg/m2 for dose level 5. Indoximod was given at 300, 600, 1000, 2000, and 1200 mg PO twice daily continuously for levels 1-5, respectively. Serum drug levels were measured. RESULTS Twenty-seven patients were treated, with 22 evaluable for response. DLTs included grade 3 dehydration (level 1), hypotension(level 4), mucositis (level 4) and grade 5 enterocolitis (level 2). Dose level 5 is the recommended phase II dose. The most frequent adverse events were fatigue (58.6%), anemia (51.7%), hyperglycemia (48.3%), infection (44.8%), and nausea (41.4%). There were 4 partial responses (2 breast, 1 NSCLC, 1 thymic tumor). No drug-drug interactions were noted. CONCLUSIONS Docetaxel plus indoximod was well tolerated with no increase in expected toxicities or pharmacokinetic interactions. It was active in a pretreated population of patients with metastatic solid tumors.
Collapse
Affiliation(s)
- Hatem H Soliman
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | - Tony Neuger
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - E Claire Dees
- University of North Carolina/Lineberger Cancer Center, Chapel Hill, NC
| | | | - Hyo Han
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | - Susan Minton
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | | | | | - Scott Antonia
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
39
|
Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, Apetoh L, Aranda F, Barnaba V, Bloy N, Bracci L, Breckpot K, Brough D, Buqué A, Castro MG, Cirone M, Colombo MI, Cremer I, Demaria S, Dini L, Eliopoulos AG, Faggioni A, Formenti SC, Fučíková J, Gabriele L, Gaipl US, Galon J, Garg A, Ghiringhelli F, Giese NA, Guo ZS, Hemminki A, Herrmann M, Hodge JW, Holdenrieder S, Honeychurch J, Hu HM, Huang X, Illidge TM, Kono K, Korbelik M, Krysko DV, Loi S, Lowenstein PR, Lugli E, Ma Y, Madeo F, Manfredi AA, Martins I, Mavilio D, Menger L, Merendino N, Michaud M, Mignot G, Mossman KL, Multhoff G, Oehler R, Palombo F, Panaretakis T, Pol J, Proietti E, Ricci JE, Riganti C, Rovere-Querini P, Rubartelli A, Sistigu A, Smyth MJ, Sonnemann J, Spisek R, Stagg J, Sukkurwala AQ, Tartour E, Thorburn A, Thorne SH, Vandenabeele P, Velotti F, Workenhe ST, Yang H, Zong WX, Zitvogel L, Kroemer G, Galluzzi L. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology 2014; 3:e955691. [PMID: 25941621 PMCID: PMC4292729 DOI: 10.4161/21624011.2014.955691] [Citation(s) in RCA: 617] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/04/2014] [Indexed: 02/07/2023] Open
Abstract
Apoptotic cells have long been considered as intrinsically tolerogenic or unable to elicit immune responses specific for dead cell-associated antigens. However, multiple stimuli can trigger a functionally peculiar type of apoptotic demise that does not go unnoticed by the adaptive arm of the immune system, which we named "immunogenic cell death" (ICD). ICD is preceded or accompanied by the emission of a series of immunostimulatory damage-associated molecular patterns (DAMPs) in a precise spatiotemporal configuration. Several anticancer agents that have been successfully employed in the clinic for decades, including various chemotherapeutics and radiotherapy, can elicit ICD. Moreover, defects in the components that underlie the capacity of the immune system to perceive cell death as immunogenic negatively influence disease outcome among cancer patients treated with ICD inducers. Thus, ICD has profound clinical and therapeutic implications. Unfortunately, the gold-standard approach to detect ICD relies on vaccination experiments involving immunocompetent murine models and syngeneic cancer cells, an approach that is incompatible with large screening campaigns. Here, we outline strategies conceived to detect surrogate markers of ICD in vitro and to screen large chemical libraries for putative ICD inducers, based on a high-content, high-throughput platform that we recently developed. Such a platform allows for the detection of multiple DAMPs, like cell surface-exposed calreticulin, extracellular ATP and high mobility group box 1 (HMGB1), and/or the processes that underlie their emission, such as endoplasmic reticulum stress, autophagy and necrotic plasma membrane permeabilization. We surmise that this technology will facilitate the development of next-generation anticancer regimens, which kill malignant cells and simultaneously convert them into a cancer-specific therapeutic vaccine.
Collapse
Key Words
- APC, antigen-presenting cell
- ATF6, activating transcription factor 6
- ATP release
- BAK1, BCL2-antagonist/killer 1
- BAX, BCL2-associated X protein
- BCL2, B-cell CLL/lymphoma 2 protein
- CALR, calreticulin
- CTL, cytotoxic T lymphocyte
- DAMP, damage-associated molecular pattern
- DAPI, 4′,6-diamidino-2-phenylindole
- DiOC6(3), 3,3′-dihexyloxacarbocyanine iodide
- EIF2A, eukaryotic translation initiation factor 2A
- ER, endoplasmic reticulum
- FLT3LG, fms-related tyrosine kinase 3 ligand
- G3BP1, GTPase activating protein (SH3 domain) binding protein 1
- GFP, green fluorescent protein
- H2B, histone 2B
- HMGB1
- HMGB1, high mobility group box 1
- HSP, heat shock protein
- HSV-1, herpes simplex virus type I
- ICD, immunogenic cell death
- IFN, interferon
- IL, interleukin
- MOMP, mitochondrial outer membrane permeabilization
- PDIA3, protein disulfide isomerase family A
- PI, propidium iodide
- RFP, red fluorescent protein
- TLR, Toll-like receptor
- XBP1, X-box binding protein 1
- autophagy
- calreticulin
- endoplasmic reticulum stress
- immunotherapy
- member 3
- Δψm, mitochondrial transmembrane potential
Collapse
Affiliation(s)
- Oliver Kepp
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Laura Senovilla
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
- INSERM; U1015; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
| | - Erika Vacchelli
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Sandy Adjemian
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Molecular Cell Biology Laboratory; Department of Immunology; Institute of Biomedical Sciences; University of São Paulo; São Paulo, Brazil
| | - Patrizia Agostinis
- Cell Death Research and Therapy (CDRT) Laboratory; Department of Cellular and Molecular Medicine; University of Leuven; Leuven, Belgium
| | - Lionel Apetoh
- INSERM; UMR866; Dijon, France
- Centre Georges François Leclerc; Dijon, France
- Université de Bourgogne; Dijon, France
| | - Fernando Aranda
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Vincenzo Barnaba
- Departement of Internal Medicine and Medical Sciences; University of Rome La Sapienza; Rome, Italy
- Istituto Pasteur; Fondazione Cenci Bolognetti; Rome, Italy
| | - Norma Bloy
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Laura Bracci
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy (LMCT); Department of Biomedical Sciences Medical School of the Free University of Brussels (VUB); Jette, Belgium
| | - David Brough
- Faculty of Life Sciences; University of Manchester; Manchester, UK
| | - Aitziber Buqué
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Maria G. Castro
- Department of Neurosurgery and Cell and Developmental Biology; University of Michigan School of Medicine; Ann Arbor, MI USA
| | - Mara Cirone
- Department of Experimental Medicine; University of Rome La Sapienza; Rome, Italy
| | - Maria I. Colombo
- Laboratorio de Biología Celular y Molecular; Instituto de Histología y Embriología (IHEM); Facultad de Ciencias Médicas; Universidad Nacional de Cuyo; CONICET; Mendoza, Argentina
| | - Isabelle Cremer
- INSERM; U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Equipe 13; Center de Recherche des Cordeliers; Paris, France
| | - Sandra Demaria
- Department of Pathology; New York University School of Medicine; New York, NY USA
| | - Luciana Dini
- Department of Biological and Environmental Science and Technology (DiSTeBA); University of Salento; Lecce, Italy
| | - Aristides G. Eliopoulos
- Molecular and Cellular Biology Laboratory; Division of Basic Sciences; University of Crete Medical School; Heraklion, Greece
- Institute of Molecular Biology and Biotechnology; Foundation of Research and Technology - Hellas; Heraklion, Greece
| | - Alberto Faggioni
- Department of Experimental Medicine; University of Rome La Sapienza; Rome, Italy
| | - Silvia C. Formenti
- Department of Radiation Oncology; NewYork University School of Medicine and Langone Medical Center; New York, NY USA
| | - Jitka Fučíková
- Department of Immunology; 2 Faculty of Medicine and University Hospital Motol, Charles University; Prague, Czech Republic
- Sotio; Prague, Czech Republic
| | - Lucia Gabriele
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Udo S. Gaipl
- Department of Radiation Oncology; University Hospital Erlangen; University of Erlangen-Nürnberg; Erlangen, Germany
| | - Jérôme Galon
- INSERM; U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Laboratory of Integrative Cancer Immunology; Center de Recherche des Cordeliers; Paris, France
| | - Abhishek Garg
- Cell Death Research and Therapy (CDRT) Laboratory; Department of Cellular and Molecular Medicine; University of Leuven; Leuven, Belgium
| | - François Ghiringhelli
- INSERM; UMR866; Dijon, France
- Centre Georges François Leclerc; Dijon, France
- Université de Bourgogne; Dijon, France
| | - Nathalia A. Giese
- European Pancreas Center; Department of Surgery; University Hospital Heidelberg; Heidelberg, Germany
| | - Zong Sheng Guo
- Department of Surgery; University of Pittsburgh; Pittsburgh, PA USA
| | - Akseli Hemminki
- Cancer Gene Therapy Group; Transplantation laboratory; Haartman Institute; University of Helsinki; Helsinki, Finland
| | - Martin Herrmann
- Department of Internal Medicine 3; University of Erlangen-Nuremberg; Erlangen, Germany
| | - James W. Hodge
- Laboratory of Tumor Immunology and Biology; Center for Cancer Research; National Cancer Institute (NCI), National Institutes of Health (NIH); Bethesda, MD USA
| | - Stefan Holdenrieder
- Institute of Clinical Chemistry and Clinical Pharmacology; University Hospital Bonn; Bonn, Germany
| | - Jamie Honeychurch
- Faculty of Medical and Human Sciences, Institute of Cancer Studies; Manchester Academic Health Sciences Center; University of Manchester; Manchester, UK
| | - Hong-Min Hu
- Cancer Research and Biotherapy Center; Second Affiliated Hospital of Southeast University; Nanjing, China
- Laboratory of Cancer Immunobiology; Earle A. Chiles Research Institute; Providence Portland Medical Center; Portland, OR USA
| | - Xing Huang
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Tim M. Illidge
- Faculty of Medical and Human Sciences, Institute of Cancer Studies; Manchester Academic Health Sciences Center; University of Manchester; Manchester, UK
| | - Koji Kono
- Department of Surgery; National University of Singapore; Singapore, Singapore
- Cancer Science Institute of Singapore; National University of Singapore; Singapore, Singapore
| | | | - Dmitri V. Krysko
- VIB Inflammation Research Center; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
| | - Sherene Loi
- Division of Cancer Medicine and Division of Research; Peter MacCallum Cancer Center; East Melbourne; Victoria, Australia
| | - Pedro R. Lowenstein
- Department of Neurosurgery and Cell and Developmental Biology; University of Michigan School of Medicine; Ann Arbor, MI USA
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology; Humanitas Clinical and Research Center; Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan; Rozzano, Italy
| | - Yuting Ma
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Frank Madeo
- Institute of Molecular Biosciences; University of Graz; Graz, Austria
| | - Angelo A. Manfredi
- University Vita-Salute San Raffaele; Milano, Italy
- San Raffaele Scientific Institute; Milano, Italy
| | - Isabelle Martins
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1030; Villejuif, France
- Faculté de Médecine; Université Paris-Sud/Paris XI; Kremlin-Bicêtre, France
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology; Humanitas Clinical and Research Center; Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan; Rozzano, Italy
| | - Laurie Menger
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Cancer Immunology Unit, Research Department of Haematology; University College London (UCL) Cancer Institute; London, UK
| | - Nicolò Merendino
- Department of Ecological and Biological Sciences (DEB), Tuscia University; Viterbo, Italy
| | - Michael Michaud
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Gregoire Mignot
- Cellular and Molecular Immunology and Endocrinology, Oniris; Nantes, France
| | - Karen L. Mossman
- Department of Pathology and Molecular Medicine; McMaster Immunology Research Center; Hamilton, Canada
- Institute for Infectious Disease Research; McMaster University; Hamilton, Canada
| | - Gabriele Multhoff
- Department of Radiation Oncology; Klinikum rechts der Isar; Technical University of Munich; Munich, Germany
| | - Rudolf Oehler
- Comprehensive Cancer Center; Medical University of Vienna; Vienna, Austria
| | - Fabio Palombo
- Departement of Internal Medicine and Medical Sciences; University of Rome La Sapienza; Rome, Italy
- Istituto Pasteur; Fondazione Cenci Bolognetti; Rome, Italy
| | | | - Jonathan Pol
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Enrico Proietti
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Jean-Ehrland Ricci
- INSERM; U1065; Nice, France
- Equipe “Contrôle Métabolique des Morts Cellulaires,” Center Méditerranéen de Médecine Moléculaire (C3M); Nice, France
- Faculté de Médecine; Université de Nice Sophia Antipolis; Nice, France
- Centre Hospitalier Universitaire de Nice; Nice, France
| | - Chiara Riganti
- Department of Oncology and Subalpine Center for Research and Experimental Medicine (CeRMS); University of Turin; Turin, Italy
| | - Patrizia Rovere-Querini
- University Vita-Salute San Raffaele; Milano, Italy
- San Raffaele Scientific Institute; Milano, Italy
| | - Anna Rubartelli
- Cell Biology Unit; Azienda Ospedaliera Universitaria San Martino; Istituto Nazionale per la Ricerca sul Cancro; Genova, Italy
| | | | - Mark J. Smyth
- Immunology in Cancer and Infection Laboratory; QIMR Berghofer Medical Research Institute; Herston, Australia
- School of Medicine, University of Queensland; Herston, Australia
| | - Juergen Sonnemann
- Department of Pediatric Haematology and Oncology; Jena University Hospital, Children's Clinic; Jena, Germany
| | - Radek Spisek
- Department of Immunology; 2 Faculty of Medicine and University Hospital Motol, Charles University; Prague, Czech Republic
- Sotio; Prague, Czech Republic
| | - John Stagg
- Centre de Recherche du Center Hospitalier de l’Université de Montréal; Faculté de Pharmacie, Université de Montréal; Montréal, Canada
| | - Abdul Qader Sukkurwala
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Department of Pathology, Dow International Medical College; Dow University of Health Sciences; Karachi, Pakistan
| | - Eric Tartour
- INSERM; U970; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
| | - Andrew Thorburn
- Department of Pharmacology; University of Colorado School of Medicine; Aurora, CO USA
| | | | - Peter Vandenabeele
- VIB Inflammation Research Center; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
- Methusalem Program; Ghent University; Ghent, Belgium
| | - Francesca Velotti
- Department of Ecological and Biological Sciences (DEB), Tuscia University; Viterbo, Italy
| | - Samuel T. Workenhe
- Department of Pathology and Molecular Medicine; McMaster Immunology Research Center; Hamilton, Canada
- Institute for Infectious Disease Research; McMaster University; Hamilton, Canada
| | - Haining Yang
- University of Hawaii Cancer Center; Honolulu, HI USA
| | - Wei-Xing Zong
- Department of Molecular Genetics and Microbiology; Stony Brook University; Stony Brook, NY USA
| | - Laurence Zitvogel
- INSERM; U1015; Villejuif, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Centre d’Investigation Clinique Biothérapie 507 (CICBT507); Gustave Roussy Cancer Campus; Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
40
|
Encapsulated cells expressing a chemotherapeutic activating enzyme allow the targeting of subtoxic chemotherapy and are safe and efficacious: data from two clinical trials in pancreatic cancer. Pharmaceutics 2014; 6:447-66. [PMID: 25116885 PMCID: PMC4190529 DOI: 10.3390/pharmaceutics6030447] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 07/02/2014] [Accepted: 07/30/2014] [Indexed: 02/07/2023] Open
Abstract
Despite progress in the treatment of pancreatic cancer, there is still a need for improved therapies. In this manuscript, we report clinical experience with a new therapy for the treatment of pancreatic cancer involving the implantation of encapsulated cells over-expressing a cytochrome P450 enzyme followed by subsequent low-dose ifosfamide administrations as a means to target activated ifosfamide to the tumor. The safety and efficacy of the angiographic instillation of encapsulated allogeneic cells overexpressing cytochrome P450 in combination with low-dose systemic ifosfamide administration has now been evaluated in 27 patients in total. These patients were successfully treated in four centers by three different interventional radiologists, arguing strongly that the treatment can be successfully used in different centers. The safety of the intra-arterial delivery of the capsules and the lack of evidence that the patients developed an inflammatory or immune response to the encapsulated cells or encapsulation material was shown in all 27 patients. The ifosfamide dose of 1 g/m2/day used in the first trial was well tolerated by all patients. In contrast, the ifosfamide dose of 2 g/m2/day used in the second trial was poorly tolerated in most patients. Since the median survival in the first trial was 40 weeks and only 33 weeks in the second trial, this strongly suggests that there is no survival benefit to increasing the dose of ifosfamide, and indeed, a lower dose is beneficial for quality of life and the lack of side effects. This is supported by the one-year survival rate in the first trial being 38%, whilst that in the second trial was only 23%. However, taking the data from both trials together, a total of nine of the 27 patients were alive after one year, and two of these nine patients were alive for two years or more.
Collapse
|