1
|
Kim GM, Kim S, Lee MA, Byun MS, Choi D, Yang SH, Woo J, Sung YC, Shin EC, Park SH, Kim TW, Sohn J. GX-I7, a long-acting IL-7, safely and effectively increased peripheral CD8 +/CD4 + T cells and TILs in patients with locally advanced or metastatic solid tumours. Br J Cancer 2025:10.1038/s41416-025-03069-3. [PMID: 40490502 DOI: 10.1038/s41416-025-03069-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 05/07/2025] [Accepted: 05/20/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND GX-I7 (rhIL-7-hyFc, efineptakin alfa) is a hybrid Fc-fused long-acting interleukin-7 (IL-7) with the aim of correcting T-cell deficiency, thereby strengthening the immune response to fight against cancer. This Phase 1b, dose-escalation study was designed to assess the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of GX-I7 in patients with locally advanced or metastatic solid tumours. METHODS This study, conducted in patients with advanced solid tumours at three hospitals in Korea, involved intramuscular GX-I7 administration across eight dose levels (60-1700 µg/kg) in 3- and 6-week cohorts. A dose-expansion phase at 720 and 1200 µg/kg further assessed GX-I7's safety and efficacy. RESULTS Anti-tumour responses showed either stable disease (SD) or disease progression (PD). GX-I7 demonstrated dose-dependent increases in the maximum serum concentration (Cmax) and area under the curve up to the last measurable concentration (AUClast). In addition, a dose-dependent increase in circulating CD8+/CD4+ T cells was observed. In five patients who consented for biopsy, a statistically significant increase in tumour-infiltrating lymphocytes (TILs) followed GX-I7 treatment. DISCUSSION Findings suggest GX-I7 is a safe T cell-amplifying agent with peripheral immune activation. Ongoing studies are exploring its ability to enhance immune responses in peripheral immune cells and tumour cells when combined with other anti-cancer agents. CLINICAL TRIAL REGISTRATION NCT03478995.
Collapse
Affiliation(s)
- Gun Min Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Sojeong Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Myung Ah Lee
- Division of Medical Oncology, Seoul St, Mary's Hospital, The catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea.
| | - Tae Won Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.
| | - Joohyuk Sohn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
2
|
Ohri N, Bodner WR, Kabarriti R, Shankar V, Gucalp R, Cheng H, Halmos B. Randomized Evaluation of the PET-Adjusted IMRT for NSCLC Trial (REPAINT). Int J Radiat Oncol Biol Phys 2025; 122:365-373. [PMID: 39862898 DOI: 10.1016/j.ijrobp.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/17/2024] [Accepted: 01/12/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Standard radiation therapy (RT) for locally advanced nonsmall cell lung cancer (LA-NSCLC) employs a uniform dose of approximately 60 Gy. Recent trials demonstrated that RT dose escalation may not improve outcomes and may cause added toxicity. We previously performed a single-arm trial testing a personalized, risk-adapted, and deintensified RT strategy. We now report findings from a randomized trial testing this novel approach. METHODS AND MATERIALS Patients with LA-NSCLC with Eastern Cooperative Oncology Group performance status 0-2 were eligible for this trial. Metabolic tumor volume for each pulmonary tumor and involved lymph node was calculated using fludeoxyglucose PET. Participants were randomly assigned 1:1 to receive standard RT (60 Gy in 30 fractions delivered to pulmonary tumors and involved lymph nodes) versus dose-painted RT (55 Gy delivered to tumors and lymph nodes with metabolic volume exceeding 20 cm3 and 44-48 Gy to other lesions, all in 20 fractions). Concurrent chemotherapy and standard adjuvant therapy were given in both arms. The primary objective was to characterize patient-reported outcomes using Patient-Reported Outcomes Version of the Common Terminology Criteria for Adverse Events. Secondary objectives included comparing outcomes between study arms. RESULTS Fifty patients were enrolled. The most common grade 3 patient-reported adverse events within 90 days of RT completion were dysphagia (38%), fatigue (38%), cough (32%), and wheezing (28%). The median progression-free survival duration is 18 months, and the median overall survival duration is 42 months. Progression-free survival and overall survival rates are similar across study arms (logrank P = .562 and .765, respectively). There have been 3 cases of in-field disease progression, with 1 in the control arm and 2 in the dose-painted arm. Grade 3-4 lymphopenia was reduced with dose-painted RT (48% vs 81%, chi-square P = .012). CONCLUSION High-grade patient-reported toxicity in patients with LA-NSCLC who are treated with concurrent chemoradiotherapy is common. We found no evidence that risk-adapted RT de-escalation compromises clinical outcomes. Follow-up studies testing the ability of this approach to improve the safety profile of chemoradiotherapy are warranted.
Collapse
Affiliation(s)
- Nitin Ohri
- Department of Radiation Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York.
| | - William R Bodner
- Department of Radiation Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York
| | - Rafi Kabarriti
- Department of Radiation Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York
| | - Viswanathan Shankar
- Department of Epidemiology and Population Health, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York
| | - Rasim Gucalp
- Department of Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York
| | - Haiying Cheng
- Department of Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York
| | - Balazs Halmos
- Department of Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York
| |
Collapse
|
3
|
Han L, Huang B, Li L, Xu B, Yang Y, Zhao L, Wang Z, Zhang C, Gao Q. Utility of the neutrophil-to-lymphocyte ratio and the ratio of neutrophil-to-lymphocyte ratio after and before adverse events for differential diagnosis of immune-related adverse events and bacterial infections in cancer patients treated with PD-(L)1 inhibitors. J Leukoc Biol 2025; 117:qiaf029. [PMID: 40083232 DOI: 10.1093/jleuko/qiaf029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/25/2024] [Accepted: 03/13/2025] [Indexed: 03/16/2025] Open
Abstract
Differential diagnosis of immune-related adverse events (irAEs) or bacterial infections is sometimes very difficult in cancer patients undergoing treatment with PD-(L)1 inhibitors. This study aimed to assess the effectiveness of the neutrophil-to-lymphocyte ratio (NLR) in distinguishing between irAEs and bacterial infections in cancer patients receiving PD-(L)1 inhibitors. We conducted a retrospective analysis of cancer patients who received at least 1 dose of PD-(L)1 inhibitors at Affiliated Cancer Hospital of Zhengzhou University from 2018 to 2023. We compared the changes in peripheral blood cell counts before and after the occurrence of adverse events, as well as the ratios of the NLR that were closest after the occurrence of adverse events (post-NLR) to the NLR that were closest before the occurrence of adverse events (pre-NLR). Among the 4173 patients who were administered PD-(L)1 inhibitors, 217 individuals experienced a total of 249 irAEs, while 256 patients were diagnosed with 257 bacterial infections. The post-NLR increased significantly compared with pre-NLR in patients with bacterial infection (P < 0.001), while the post-NLR had smaller increase compared with pre-NLR in patients sufffering irAEs (P < 0.001). Notably, the NLR was significantly higher in patients with bacterial infection compared with those with irAEs (P < 0.001). Furthermore, the post-NLR/pre-NLR ratio was higher in the bacterial infection group than in the irAEs group (P < 0.001). The NLR along with the post-NLR/pre-NLR ratio could serve as valuable diagnostic indicators for irAEs and bacterial infections in cancer patients undergoing treatment with PD-(L)1 inhibitors.
Collapse
Affiliation(s)
- Lu Han
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Beibei Huang
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Linlin Li
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Benling Xu
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Yonghao Yang
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Lingdi Zhao
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Zibing Wang
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Chaoji Zhang
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Quanli Gao
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| |
Collapse
|
4
|
Bracamonte-Baran W, Kim ST. The Current and Future of Biomarkers of Immune Related Adverse Events. Immunol Allergy Clin North Am 2025; 45:223-249. [PMID: 40287170 DOI: 10.1016/j.iac.2025.01.010] [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] [Indexed: 04/29/2025]
Abstract
With their groundbreaking clinical responses, immune checkpoint inhibitors (ICIs) have ushered in a new chapter in cancer therapeutics. However, they are often associated with life-threatening or organ-threatening autoimmune/autoinflammatory phenomena, collectively termed immune-related adverse events (irAEs). In this review, we will first describe the mechanisms of action of ICIs as well as irAEs. Next, we will review biomarkers for predicting the development of irAEs or stratifying risks.
Collapse
Affiliation(s)
- William Bracamonte-Baran
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA
| | - Sang T Kim
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA.
| |
Collapse
|
5
|
Natori K, Igeta M, Morimoto T, Nagahashi M, Akashi-Tanaka S, Daimon T, Miyoshi Y. Development and internal validation of a predictive model of overall and progression-free survival in eribulin-treated patients with breast cancer based on baseline peripheral blood parameters. Breast Cancer 2025; 32:500-511. [PMID: 39979692 PMCID: PMC11993485 DOI: 10.1007/s12282-025-01678-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/01/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Immune and inflammatory blood parameters have been reported as biomarkers for treatment efficacy. This study aimed to establish a predictive model that includes blood parameters for patients with metastatic breast cancer treated with eribulin. METHODS A total of 297 patients were enrolled, and their baseline neutrophil-to-lymphocyte ratio, absolute lymphocyte count (ALC), platelet-to-lymphocyte ratio (PLR), prognostic nutritional index (PNI), lymphocyte-to-monocyte ratio (LMR), lactate dehydrogenase (LDH), C-reactive protein (CRP), and clinical data were retrospectively collected. RESULTS We constructed nomograms to predict overall survival (OS) and progression-free survival (PFS) using blood parameters, including clinical factors. For OS, menopausal status, hormone receptor status, HER2 status, de novo or recurrent, metastatic site, treatment line, ALC, PLR, PNI, LMR, LDH, and CRP were selected to predict the model. We used menopausal status, hormone receptor status, HER2 status, treatment line, PLR, LMR, LDH, and CRP to predict PFS. Both the OS and PFS of patients according to the risk scores were significantly different (p < 0.001). The optimism-corrected C-indices of the nomograms for OS and PFS were 0.680 and 0.622, respectively. The mean time-dependent area under the receiver operating curve values for OS at 1, 2, and 3 years were 0.752, 0.761, and 0.784, respectively, and for PFS at 3, 6, and 12 months were 0.660, 0.661, and 0.650, respectively. CONCLUSION Nomograms incorporating peripheral blood parameters may improve the accuracy of predicting OS and PFS in patients treated with eribulin. Our prediction model may help decision-making for breast cancer patients who are considering eribulin treatment.
Collapse
Affiliation(s)
- Keiko Natori
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
- Department of Breast Surgery, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Masataka Igeta
- Department of Biostatistics, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Takashi Morimoto
- Department of Breast Surgery, Yao Municipal Hospital, 1-3-1 Ryuge-Cho, Yao, Osaka, 581-0069, Japan
| | - Masayuki Nagahashi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Sadako Akashi-Tanaka
- Department of Breast Surgery, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Takashi Daimon
- Department of Biostatistics, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Yasuo Miyoshi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan.
| |
Collapse
|
6
|
Zhao XR, Fang H, Jing H, Zhong QZ, Wu HF, Hou XR, Dong LH, Zhong YH, Jin J, Zhao LN, Wang XH, Yang WF, Tie J, Lu YF, Sun GY, Wang DQ, Tang Y, Qi SN, Song YW, Liu YP, Tang Y, Lu NN, Chen B, Zhang WW, Zhai YR, Hu SY, Zhang J, Li YX, Zhang N, Wang SL. Longitudinal Analyses and Predictive Factors of Radiation-Induced Lymphopenia After Postmastectomy Hypofractionated Radiation Therapy for Breast Cancer: A Pooled Cohort Study of 2 Prospective Trials. Adv Radiat Oncol 2025; 10:101750. [PMID: 40241739 PMCID: PMC12002827 DOI: 10.1016/j.adro.2025.101750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/18/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose Radiation-induced lymphopenia (RIL) correlates with poor prognoses in solid tumors. This study aimed to investigate the post-radiation therapy (RT) longitudinal lymphocyte changes and the impact of different RT techniques on RIL in breast cancer patients. Methods and Materials We prospectively assessed 607 breast cancer patients who received hypofractionated postmastectomy RT in 8 hospitals. Radiation therapy techniques included integrated photon-based intensity modulated technique (integrated RT) and a combination of photon irradiation of supraclavicular nodes and electron irradiation of the chest wall and/or the internal mammary node (hybrid RT). Peripheral lymphocyte counts (PLC) were determined before RT, weekly during RT, at 1 and 2 weeks, 3 and 6 months post-RT, and then every 6 months. The primary outcome was the nadir PLC during RT, for which associated factors were analyzed. Univariate, multivariable linear regression and propensity score matching analyses were performed to evaluate the effect of different RT techniques on nadir PLC. Results During RT, 121 (19.9%) patients had grade ≥3 RIL with a nadir PLC of 0.75 ± 0.33 × 109/L. The PLC started to recover at 1 week and reached pre-RT levels 1 year after RT and higher than pre-RT levels 2 years later. Multivariate analysis identified young age, low body mass index, radiation therapy targets involving multiple regions, integrated RT, and low pre-radiation therapy PLC as independent risk factors for nadir PLC (P < .005). The PLC at each time point during and after radiation therapy was lower in patients receiving integrated RT than in those receiving hybrid RT (P < .05). Before and after propensity score matching, integrated RT was significantly associated with lower nadir PLC after adjusting for radiation therapy targets and age (P < .001). Conclusions Breast cancer patients had prolonged lymphopenia post-RT. Integrated RT increased the risk of RIL and adversely affected recovery. Therefore, an appropriate RT technique should be considered to minimize RIL.
Collapse
Affiliation(s)
- Xu-Ran Zhao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hui Fang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hao Jing
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qiu-Zi Zhong
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, P.R. China
| | - Hong-Fen Wu
- Department of Radiation Oncology, JILIN Cancer Hospital, Changchun, China
| | - Xiao-Rong Hou
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Hua Dong
- Department of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Ya-Hua Zhong
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, China
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Li-Na Zhao
- Department of Radiation Oncology, Xijing Hospital, The First Affiliated Hospital of Fourth Military Medical University, Xi'an, China
| | - Xiao-Hong Wang
- Department of Radiochemotherapy, Tangshan People's Hospital, Tangshan, China
| | - Wei-Fang Yang
- Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, China
| | - Jian Tie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yu-Fei Lu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Guang-Yi Sun
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dan-Qiong Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu Tang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Nan Qi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yong-Wen Song
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yue-Ping Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuan Tang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ning-Ning Lu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bo Chen
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wen-Wen Zhang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yi-Rui Zhai
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shang-Ying Hu
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Zhang
- Department of Radiation Oncology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ye-Xiong Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Department of Radiation Oncology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Na Zhang
- Department of Radiation Oncology, Cancer Hospital of Dalian University of Technology,Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Shu-Lian Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
7
|
Braghieri L, Gharaibeh A, Nkashama L, Abushouk A, Abushawer O, Mehdizadeh‐Shrifi A, Honnekeri B, Calabrese C, Menon V, Funchain P, Collier P, Sadler D, Moudgil R. Long-term cardiovascular outcomes of immune checkpoint inhibitor-related myocarditis: A large single-centre analysis. ESC Heart Fail 2025; 12:1237-1245. [PMID: 39482568 PMCID: PMC11911570 DOI: 10.1002/ehf2.15131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/11/2024] [Accepted: 10/03/2024] [Indexed: 11/03/2024] Open
Abstract
AIMS Immune checkpoint inhibitors (ICI) are the cornerstone of modern oncology; however, side effects such as ICI-related myocarditis (irM) can be fatal. Recently, Bonaca proposed criteria for irM; however, it is unknown if they correlate well with cardiovascular (CV) ICI-related adverse events. Additionally, whether incident irM portends worse long-term CV outcomes remains unclear. We aimed to determine the incidence of long-term CV comorbidities and CV mortality among irM patients. PATIENTS AND METHODS The ICI-related adverse event (irAE) registry was queried to identify irM patients by using Bonaca criteria. Random controls were selected after excluding patients with other concomitant irAEs. Patients' demographics, comorbidities and myocarditis presenting features were gathered. Outcomes included 2-year freedom from CV comorbidities (composite of atrial fibrillation, stroke, myocardial infarction and heart failure) and freedom from CV death. IrM was treated as a time-varying covariate. RESULTS Seventy-six patients developed irM at a median of 167 days (mean age 69, 63.2% male, 47% lung cancer). Majority of patients had new wall motion abnormalities or EKG changes on presentation. Mean LVEF was 43%, median peak TnT was 0.81, and median NTproBNP was 2057 at irM onset. Two-year freedom from CV comorbidities (67% vs 86.8%, P < 0.001) and death (93.4% vs 99.3%, P = 0.003) was lower among irM patients. Incident irM was an independent predictor of CV death (HR 8.28, P = 0.048), but not CV comorbidities (HR 2.21, P = 0.080). CONCLUSIONS This is the largest case-control study on irM highlighting worse long-term CV outcomes. Future studies are needed to establish appropriate therapeutic strategies and efficient screening strategies for irM survivors.
Collapse
Affiliation(s)
- Lorenzo Braghieri
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Ahmad Gharaibeh
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Lubika Nkashama
- Department of Internal MedicineWashU/Barnes‐Jewish HospitalSt. LouisMissouriUSA
| | | | - Osama Abushawer
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | | | - Bianca Honnekeri
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Cassandra Calabrese
- Department of Rheumatologic and Immunologic DiseaseCleveland Clinic FoundationClevelandOhioUSA
| | - Venu Menon
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationClevelandOhioUSA
| | - Pauline Funchain
- Department of Hematology & OncologyTaussig Cancer Center, Cleveland ClinicClevelandOhioUSA
| | - Patrick Collier
- Department of Cardiovascular Medicine, Division of Cardiac Imaging, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationClevelandOhioUSA
| | - Diego Sadler
- Department of Cardiovascular Medicine, Division of Cardiac Imaging, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationWestonFloridaUSA
| | - Rohit Moudgil
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationClevelandOhioUSA
| |
Collapse
|
8
|
Bernardo D, Murugan NJ, Voutsadakis IA. Pre-Treatment Peripheral Blood Parameters as Prognostic Biomarkers in Cancer Patients Receiving Immune Checkpoint Inhibitors. Int J Hematol Oncol Stem Cell Res 2025; 19:7-16. [PMID: 40421401 PMCID: PMC12103826 DOI: 10.18502/ijhoscr.v19i1.17819] [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: 03/07/2023] [Accepted: 07/01/2024] [Indexed: 05/28/2025] Open
Abstract
Background: Immune checkpoint inhibitors have significantly improved outcomes in select cancers; however, not all patients respond to these therapies, and the duration of the response varies among responders. Markers predictive of the response to immunotherapy, such as PD-L1 expression determined by immunohistochemical staining of tumor sections and microsatellite status, have been identified. Some of these are used in companion diagnostics approved for clinical practice. Additional easy-to-use biomarkers may help clinicians to predict the efficacy of these drugs in individual patients. Materials and Methods: A retrospective review of the medical records of patients with metastatic cancer treated with immune checkpoint inhibitors in our cancer center was performed to identify the clinical and hematologic parameters associated with survival outcomes. Results: Among the 163 patients included in the study, most had lung cancer, followed by kidney cancer, melanoma, and bladder cancer. Most patients (61.3%) were male and had good performance status. Nivolumab and pembrolizumab were immune checkpoint inhibitors utilized in 85.9% of cases. Age, sex, and primary cancer type were not associated with survival outcomes. Among the peripheral blood parameters evaluated, lymphocytopenia was the strongest predictor of adverse survival outcomes in univariate analysis and the only clinical or hematologic biomarker that retained significance for overall survival (OS) prediction in multivariate analysis. Conclusion: Among the clinical and hematologic parameters routinely used in the clinic, a lymphocyte count below 1 x 109/ L was predictive of adverse OS in patients with metastatic cancers receiving immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | | | - Ioannis A. Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, Ontario, Canada
- Department of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| |
Collapse
|
9
|
Du Y, Zhang Y, Zhao W, Zhang Y, Su F, Zhang X, Li W, Hu W, Li Y, Zhao J. Predictive value of near-term prediction models for severe immune-related adverse events in malignant tumor PD-1 inhibitor therapy. Hum Vaccin Immunother 2024; 20:2398309. [PMID: 39267589 PMCID: PMC11404634 DOI: 10.1080/21645515.2024.2398309] [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: 06/12/2024] [Revised: 08/13/2024] [Accepted: 08/27/2024] [Indexed: 09/17/2024] Open
Abstract
Immune-related adverse events (irAEs) impact outcomes, with most research focusing on early prediction (baseline data), rather than near-term prediction (one cycle before the occurrence of irAEs and the current cycle). We aimed to explore the near-term predictive value of neutrophil/lymphocyte ratio (NLR), platelet/lymphocyte ratio (PLR), absolute eosinophil count (AEC) for severe irAEs induced by PD-1 inhibitors. Data were collected from tumor patients treated with PD-1 inhibitors. NLR, PLR, and AEC data were obtained from both the previous and the current cycles of irAEs occurrence. A predictive model was developed using elastic net logistic regression Cutoff values were determined using Youden's Index. The predicted results were compared with actual data using Bayesian survival analysis. A total of 138 patients were included, of whom 47 experienced grade 1-2 irAEs and 18 experienced grade 3-5 irAEs. The predictive model identified optimal α and λ through 10-fold cross-validation. The Shapiro-Wilk test, Kruskal-Wallis test and logistic regression showed that only current cycle data were meaningful. The NLR was statistically significant in predicting irAEs in the previous cycle. Both NLR and AEC were significant predictors of irAEs in the current cycle. The model achieved an area under the ROC curve (AUC) of 0.783, with a sensitivity of 77.8% and a specificity of 80.8%. A probability ≥ 0.1345 predicted severe irAEs. The model comprising NLR, AEC, and sex may predict the irAEs classification in the current cycle, offering a near-term predictive advantage over baseline models and potentially extending the duration of immunotherapy for patients.
Collapse
Affiliation(s)
- Yunyi Du
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Ying Zhang
- Department of Respiratory, Pengzhou People’s Hospital, Chengdu, Sichuan, China
| | - Wenqi Zhao
- Department of Statistics, University of Auckland, Auckland
| | - Yuexiang Zhang
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Fei Su
- Department of Oncology, Graduate of School of Changzhi Medical College, Changzhi, Shanxi, China
| | - Xiaoling Zhang
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Weiling Li
- Department of Oncology, The People’s Hospital of Jianyang City, Chengdu, Sichuan, China
| | - Wenqing Hu
- Department of Gastrointestinal Surgery, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Yongai Li
- Department of Radiology, Changzhi People’s Hospital, Changzhi, Shanxi, China
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jun Zhao
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| |
Collapse
|
10
|
Savino A, Rossi A, Fagiuoli S, Invernizzi P, Gerussi A, Viganò M. Hepatotoxicity in Cancer Immunotherapy: Diagnosis, Management, and Future Perspectives. Cancers (Basel) 2024; 17:76. [PMID: 39796705 PMCID: PMC11718971 DOI: 10.3390/cancers17010076] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Cancer immunotherapy, particularly immune checkpoint inhibitors, has positively impacted oncological treatments. Despite its effectiveness, immunotherapy is associated with immune-related adverse events (irAEs) that can affect any organ, including the liver. Hepatotoxicity primarily manifests as immune-related hepatitis and, less frequently, cholangitis. Several risk factors, such as pre-existing autoimmune and liver diseases, the type of immunotherapy, and combination regimens, play a role in immune-related hepatotoxicity (irH), although reliable predictive markers or models are still lacking. The severity of irH ranges from mild to severe cases, up to, in rare instances, acute liver failure. Management strategies require regular monitoring for early diagnosis and interventions, encompassing strict monitoring for mild cases to the permanent suspension of immunotherapy for severe forms. Corticosteroids are the backbone of treatment in moderate and high-grade damage, alone or in combination with additional immunosuppressive drugs for resistant or refractory cases. Given the relatively low number of events and the lack of dedicated prospective studies, much uncertainty remains about the optimal management of irH, especially in the most severe cases. This review presents the main features of irH, focusing on injury patterns and mechanisms, and provides an overview of the management landscape, from standard care to the latest evidence.
Collapse
Affiliation(s)
- Alberto Savino
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milano, Italy (M.V.)
- Gastroenterology, Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Alberto Rossi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milano, Italy (M.V.)
- Gastroenterology, Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Stefano Fagiuoli
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milano, Italy (M.V.)
- Gastroenterology, Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Pietro Invernizzi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milano, Italy (M.V.)
- Centre for Autoimmune Liver Diseases, Division of Gastroenterology, Fondazione IRCCS San Gerardo dei Tintori, ERN-RARE LIVER, 20900 Monza, Italy
| | - Alessio Gerussi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milano, Italy (M.V.)
- Centre for Autoimmune Liver Diseases, Division of Gastroenterology, Fondazione IRCCS San Gerardo dei Tintori, ERN-RARE LIVER, 20900 Monza, Italy
| | - Mauro Viganò
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milano, Italy (M.V.)
- Gastroenterology, Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| |
Collapse
|
11
|
Gaudian K, Koh MJ, Koh MJ, Jermain P, Khan I, Kallam D, Lee Z, Collins RR, Zwart Z, Danner M, Zwart A, Kumar D, Atkins MB, Suy S, Collins SP. Late radiation-related lymphopenia after prostate stereotactic body radiation therapy plus or minus supplemental pelvic irradiation. Front Oncol 2024; 14:1459732. [PMID: 39640284 PMCID: PMC11617573 DOI: 10.3389/fonc.2024.1459732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/11/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Prior studies suggest lymphopenia following radiation therapy may impact toxicity and cancer control. Chronic radiation-related lymphopenia (RRL) has been noted in prostate cancer patients treated with conventionally fractionated pelvic radiation therapy. The impact of utilizing hypofractionated high integral dose therapies such as stereotactic body radiation therapy (SBRT) on RRL is less well characterized. This prospective study sought to evaluate the impact of prostate SBRT plus or minus supplemental pelvic nodal radiation (PNI) on RRL. Methods Between 2012 and 2023, serial serum absolute lymphocyte counts (ALCs) were measured in 226 men treated at MedStar Georgetown with robotic SBRT using the CyberKnife® (CK) (36.25 Gy in 5 fractions) alone or CK (19.5 Gy in 3 fractions) followed by supplemental PNI using VMAT (37.5-45.0 Gy in 15-25 fractions) per an institutional protocol (IRB#: 2012-1175). Baseline ALC (k/μL) was measured 1-2 hours prior to robotic SBRT and at each follow-up appointment (1, 3, 6, 9, 12, 18, and 24 months post-treatment). Lymphopenia was graded using the CTCAEv.4: Grade 1 (0.8-1.0 k/μL), Grade 2 (0.5-0.8 k/μL), Grade 3 (0.2-0.5 k/μL) and Grade 4 (<0.2 k/μL). To compare two different treatment groups, the Wilcoxon signed-rank test was used. A p-value of < 0.05 determined statistical significance. Results Of 226 patients (SBRT alone: n = 169, SBRT + PNI: n = 57), the median age was 72 years and 45% of patients were non-white. Baseline lymphopenia was uncommon and of low grade. In the SBRT alone group, the baseline ALC of 1.7 k/μl decreased by 21% to 1.4 k/μL at 3 months and then stabilized. 38% of these men experienced lymphopenia in the two years following SBRT, however, no patient presented with Grade 3 lymphopenia. Patients who received SBRT + PNI had a lower baseline ALC (1.5 k/μl), and a significantly greater decrease in ALC relative to individual baseline value throughout the 2-year follow-up period, decreasing by 57% to 0.6 k/μL at 3 months and recovering to a 36% decrease from baseline (1.0 k/μL) at 24 months. Notably, 12% of the men treated with SBRT + PNI experienced Grade 3 lymphopenia. No patient in either cohort experienced Grade 4 lymphopenia. Discussion The low incidence of high-grade lymphopenia within this elderly patient population further supports the safety of prostate SBRT plus or minus PNI for the treatment of prostate cancer. However, RRL was more severe when PNI was utilized. The effect of SBRT and PNI on lymphocytes in prostate cancer patients could act as a model for other cancers, specifically those involving treatment with immunomodulatory agents. Future studies should focus on the clinical implications of RRL and the effects of specifically irradiating lymphoid tissues on lymphocyte biology.
Collapse
Affiliation(s)
- Kelly Gaudian
- Washington University School of Medicine in St. Louis, St. Louis, MO, United States
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
| | - Min Jung Koh
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
| | - Min Ji Koh
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
| | - Peter Jermain
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
| | - Irfan Khan
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
| | - Diya Kallam
- College of Medicine, George Washington University, Washington, DC, United States
| | - Zach Lee
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
| | - Ryan R. Collins
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
- Department of Radiation Oncology, University of South Florida (USF) Health Morsani College of Medicine, Tampa, FL, United States
| | - Zoya Zwart
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
| | - Malika Danner
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
- Department of Radiation Oncology, University of South Florida (USF) Health Morsani College of Medicine, Tampa, FL, United States
| | - Alan Zwart
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
| | - Deepak Kumar
- Biotechnology Research Institute, North Carolina Central University, Durham, NC, United States
| | - Michael B. Atkins
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States
| | - Simeng Suy
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
- Department of Radiation Oncology, University of South Florida (USF) Health Morsani College of Medicine, Tampa, FL, United States
| | - Sean P. Collins
- Department of Radiation Medicine, Georgetown University Hospital, Washington, DC, United States
- Department of Radiation Oncology, University of South Florida (USF) Health Morsani College of Medicine, Tampa, FL, United States
| |
Collapse
|
12
|
Macandog ADG, Catozzi C, Capone M, Nabinejad A, Nanaware PP, Liu S, Vinjamuri S, Stunnenberg JA, Galiè S, Jodice MG, Montani F, Armanini F, Cassano E, Madonna G, Mallardo D, Mazzi B, Pece S, Tagliamonte M, Vanella V, Barberis M, Ferrucci PF, Blank CU, Bouvier M, Andrews MC, Xu X, Santambrogio L, Segata N, Buonaguro L, Cocorocchio E, Ascierto PA, Manzo T, Nezi L. Longitudinal analysis of the gut microbiota during anti-PD-1 therapy reveals stable microbial features of response in melanoma patients. Cell Host Microbe 2024; 32:2004-2018.e9. [PMID: 39481388 PMCID: PMC11629153 DOI: 10.1016/j.chom.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024]
Abstract
Immune checkpoint inhibitors (ICIs) improve outcomes in advanced melanoma, but many patients are refractory or experience relapse. The gut microbiota modulates antitumor responses. However, inconsistent baseline predictors point to heterogeneity in responses and inadequacy of cross-sectional data. We followed patients with unresectable melanoma from baseline and during anti-PD-1 therapy, collecting fecal and blood samples that were surveyed for changes in the gut microbiota and immune markers. Varying patient responses were linked to different gut microbiota dynamics during ICI treatment. We select complete responders by their stable microbiota functions and validate them using multiple external cohorts and experimentally. We identify major histocompatibility complex class I (MHC class I)-restricted peptides derived from flagellin-related genes of Lachnospiraceae (FLach) as structural homologs of tumor-associated antigens, detect FLach-reactive CD8+ T cells in complete responders before ICI therapy, and demonstrate that FLach peptides improve antitumor immunity. These findings highlight the prognostic value of microbial functions and therapeutic potential of tumor-mimicking microbial peptides.
Collapse
Affiliation(s)
- Angeli D G Macandog
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Carlotta Catozzi
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Mariaelena Capone
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Amir Nabinejad
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Padma P Nanaware
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shujing Liu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Smita Vinjamuri
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612-7342, USA
| | - Johanna A Stunnenberg
- Netherlands Cancer Institute (NKI)-AVL, North Holland, Amsterdam 1066 CX, the Netherlands
| | - Serena Galiè
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Maria Giovanna Jodice
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Francesca Montani
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Federica Armanini
- Department of CIBIO, University of Trento, Trento, Povo 38123, Italy
| | - Ester Cassano
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Gabriele Madonna
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Domenico Mallardo
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | | | - Salvatore Pece
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Maria Tagliamonte
- Innovative Immunological Models, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Vito Vanella
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Massimo Barberis
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | | | - Christian U Blank
- Netherlands Cancer Institute (NKI)-AVL, North Holland, Amsterdam 1066 CX, the Netherlands
| | - Marlene Bouvier
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612-7342, USA
| | - Miles C Andrews
- Department of Medicine, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nicola Segata
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy; Department of CIBIO, University of Trento, Trento, Povo 38123, Italy
| | - Luigi Buonaguro
- Innovative Immunological Models, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Emilia Cocorocchio
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Teresa Manzo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin 10126, Italy
| | - Luigi Nezi
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy.
| |
Collapse
|
13
|
Prades-Sagarra E, Yaromina A, Dubois L. Understanding the impact of radiation-induced lymphopenia: Preclinical and clinical research perspectives. Clin Transl Radiat Oncol 2024; 49:100852. [PMID: 39315059 PMCID: PMC11418132 DOI: 10.1016/j.ctro.2024.100852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/26/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Immunotherapy has revolutionized the field of cancer treatment, changing the standard of care to the use of immune checkpoint inhibitors. Radiotherapy can boost anti-tumour immune responses by changing the tumour microenvironment, but it also can cause radiotherapy-induced lymphopenia (RIL), a decrease in circulating lymphocyte counts. RIL has been associated with lower survival in patients undergoing radiotherapy, and new studies have suggested that it can also affect immunotherapy outcome. To study RIL's effects and to explore mitigation treatment strategies, preclinical models closely mimicking the clinical situation are needed. State-of-the-art image-guided small animal irradiators now offer the possibility to target specific organs in small animals to induce RIL, aiding research on its molecular mechanisms and prevention. This review covers the relationship between radiotherapy and RIL, its impact on patient survival, and future directions to generate models to investigate and prevent RIL.
Collapse
Affiliation(s)
- E. Prades-Sagarra
- The M-Lab, Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - A. Yaromina
- The M-Lab, Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - L.J. Dubois
- The M-Lab, Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
14
|
Sun M, Xu G, Cai L, He S. Misdiagnosis of severe and atypical oxaliplatin‑related hypersensitivity reaction following chemotherapy combined with PD‑1 inhibitor: A case report and literature review. Oncol Lett 2024; 28:452. [PMID: 39101001 PMCID: PMC11292463 DOI: 10.3892/ol.2024.14586] [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: 05/10/2024] [Accepted: 07/12/2024] [Indexed: 08/06/2024] Open
Abstract
Although the efficacy of treatment strategies for cancer have been improving steadily over the past decade, the adverse event profile following such treatments has also become increasingly complex. The present report described the case of a 67-year-old male patient with gastric stump carcinoma with liver invasion. The patient was treated with oxaliplatin and capecitabine (CAPEOX regimen) chemotherapy, combined with the programmed cell death protein-1 (PD-1) inhibitor tislelizumab. Following treatment, the patient suffered from chills, high fever and facial flushing, followed by shock. Relevant examination results revealed severe multiple organ damage, as well as a significant elevation in IL-6 and procalcitonin (PCT) levels. Initially, the patient was diagnosed with either immune-related adverse events (irAEs) associated with cytokine release syndrome caused by tislelizumab or severe bacterial infection. However, when tislelizumab treatment was stopped and the CAPEOX chemotherapy regimen was reapplied, similar symptoms recurred. Following screening, it was finally determined that severe hypersensitivity reaction (HSR) caused by oxaliplatin was the cause underlying these symptoms. A literature review was then performed, which found that severe oxaliplatin-related HSR is rare, rendering the present case atypical. The present case exhibited no common HSR symptoms, such as cutaneous and respiratory symptoms. However, the patient suffered from serious multiple organ damage, which was misdiagnosed as irAE when oxaliplatin chemotherapy combined with the PD-1 inhibitor was administered. In addition, this apparent severe oxaliplatin-related HSR caused a significant increase in PCT levels, which was misdiagnosed as severe bacterial infection and prevented the use of glucocorticoids. This, in turn, aggravated the damage in this patient.
Collapse
Affiliation(s)
- Maoben Sun
- Department of Oncology, Binhaiwan Central Hospital of Dongguan, Dongguan, Guangdong 523000, P.R. China
- Dongguan Key Laboratory of Precision Medicine, Binhaiwan Central Hospital of Dongguan, Dongguan, Guangdong 523000, P.R. China
| | - Guihua Xu
- Dongguan Key Laboratory of Precision Medicine, Binhaiwan Central Hospital of Dongguan, Dongguan, Guangdong 523000, P.R. China
| | - Liangzhen Cai
- Department of Oncology, Binhaiwan Central Hospital of Dongguan, Dongguan, Guangdong 523000, P.R. China
| | - Shaozhong He
- Department of Oncology, Binhaiwan Central Hospital of Dongguan, Dongguan, Guangdong 523000, P.R. China
| |
Collapse
|
15
|
Nanos C, Koukourakis IM, Mulita A, Avgousti R, Kouloulias V, Zygogianni A, Koukourakis MI. Lymphopenia Induced by Different Neoadjuvant Chemo-Radiotherapy Schedules in Patients with Rectal Cancer: Bone Marrow as an Organ at Risk. Curr Oncol 2024; 31:5774-5788. [PMID: 39451733 PMCID: PMC11506586 DOI: 10.3390/curroncol31100429] [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: 08/17/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Radiotherapy (RT)-induced lymphopenia may hinder the anti-tumor immune response. Preoperative RT or chemo-RT (CRT) for locally advanced rectal cancer is a standard therapeutic approach, while immunotherapy has been approved for mismatch repair-deficient rectal tumors. We retrospectively analyzed 98 rectal adenocarcinoma patients undergoing neoadjuvant CRT with VMAT (groups A, B, C) or IMRT (group D) techniques, with four different RT schemes: group A (n = 24): 25 Gy/5 Gy/fraction plus a 0.2 Gy/fraction rectal tumor boost; group B (n = 22): 34 Gy/3.4 Gy/fraction, with a 1-week treatment break after the first five RT fractions; group C (n = 20): 46 Gy/2 Gy/fraction plus a 0.2 Gy/fraction rectal tumor boost; group D (n = 32): 45 Gy/1.8 Gy/fraction followed by 5.4 Gy/1.8 Gy/fraction to the rectal tumor. We examined the effect of the time-corrected normalized total dose (NTD-T) to the BM on lymphopenia. Groups A and B (hypofractionated RT) had significantly higher lymphocyte counts (LCs) after RT than groups C and D (p < 0.03). An inverse association between the LCs after RT and NTD-T was demonstrated (p = 0.01). An NTD-T threshold of 30 Gy delivered to 30% of the BM volume emerged as a potential constraint for RT planning, which could be successfully integrated in the RT plan. Hypofractionated and accelerated RT schemes, and BM-sparing techniques may reduce lymphocytic damage and prove critical for immuno-RT clinical trials.
Collapse
Affiliation(s)
- Christos Nanos
- Department of Radiotherapy and Oncology, Medical School, Democritus University of Thrace, University Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece; (C.N.); (A.M.)
| | - Ioannis M. Koukourakis
- Radiation Oncology Unit, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.M.K.); (A.Z.)
| | - Admir Mulita
- Department of Radiotherapy and Oncology, Medical School, Democritus University of Thrace, University Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece; (C.N.); (A.M.)
| | - Raphaela Avgousti
- Medical Physics Unit, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Vassilios Kouloulias
- Department of Clinical Radiation Oncology, Attikon Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Anna Zygogianni
- Radiation Oncology Unit, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.M.K.); (A.Z.)
| | - Michael I. Koukourakis
- Department of Radiotherapy and Oncology, Medical School, Democritus University of Thrace, University Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece; (C.N.); (A.M.)
| |
Collapse
|
16
|
Sloan AE, Winter K, Gilbert MR, Aldape K, Choi S, Wen PY, Butowski N, Iwamoto FM, Raval RR, Voloschin AD, Kamiya-Matsuoka C, Won M, Mehta MP. NRG-BN002: Phase I study of ipilimumab, nivolumab, and the combination in patients with newly diagnosed glioblastoma. Neuro Oncol 2024; 26:1628-1637. [PMID: 38874333 PMCID: PMC11376446 DOI: 10.1093/neuonc/noae058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have efficacy in several solid tumors but limited efficacy in glioblastoma (GBM). This study evaluated the safety of anti-CTLA-4 and anti-PD-1 ICIs alone or in combination in newly diagnosed GBM after completion of standard radiochemotherapy with the subsequent intent to test combinatorial ICIs in this setting. METHODS The primary endpoint was dose-limiting toxicity (DLT) for adults with unifocal, supratentorial newly diagnosed GBM after resection and chemoradiation. Ipilimumab and nivolumab were tested separately and in combination with a planned expansion cohort dependent upon DLT results. RESULTS Thirty-two patients were enrolled at 9 institutions: 6 to each DLT assessment cohort and 14 to the expansion cohort. Median age: 55 years, 67.7% male, 83.9% White. Treatment was well tolerated with 16% Grade 4 events; the combination did not have unexpectedly increased toxicity, with no Grade 5 events. One DLT was seen in each single-agent treatment; none were observed in the combination, leading to expanded accrual of the combined treatment. The median follow-up was 19.6 months. For all patients receiving combination treatment, median overall survival (OS) and progression-free survival (PFS) were 20.7 and 16.1 months, respectively. CONCLUSIONS IPI and NIVO are safe and tolerable with toxicities similar to those noted with other cancers when given in combination with adjuvant temozolomide for newly diagnosed GBM. Combination IPI + NIVO is not substantially more toxic than single agents. These results support a subsequent efficacy trial to test the combination of ICIs in Phase II/III for patients with newly diagnosed GBM. CLINICALTRIALS.GOV REGISTRATION NCT02311920.
Collapse
Affiliation(s)
- Andrew E Sloan
- Piedmont Healthcare, Atlanta, Georgia, USA
- Case Western Reserve University & Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Kathryn Winter
- NRG Oncology Statistics and Data Management Center/ACR, Philadelphia, Pennsylvania, USA
- American College of Radiology, Philadelphia, Pennsylvania, USA
| | - Mark R Gilbert
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
| | - Kenneth Aldape
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
| | - Serah Choi
- Case Western Reserve University & Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Patrick Y Wen
- Dana-Farber/Harvard Cancer Center, Boston, Massachusetts, USA
| | | | - Fabio M Iwamoto
- Columbia University Minority Underserved NCORP, New York, New York, USA
| | - Raju R Raval
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | | | | | - Minhee Won
- NRG Oncology Statistics and Data Management Center/ACR, Philadelphia, Pennsylvania, USA
- Case Western Reserve University & Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | | |
Collapse
|
17
|
Lee G, Kim DW, Smart AC, Horick NK, Eyler CE, Roberts HJ, Pathak P, Goyal L, Franses J, Heather JM, Hwang WL, Grassberger C, Klempner SJ, Drapek LC, Allen JN, Blaszkowsky LS, Parikh AR, Ryan DP, Clark JW, Hong TS, Wo JY. Hypofractionated Radiotherapy-Related Lymphopenia Is Associated With Worse Survival in Unresectable Intrahepatic Cholangiocarcinoma. Am J Clin Oncol 2024; 47:373-382. [PMID: 38767086 DOI: 10.1097/coc.0000000000001108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
OBJECTIVE The aim of this study was to evaluate the incidence of radiotherapy (RT)-related lymphopenia, its predictors, and association with survival in unresectable intrahepatic cholangiocarcinoma (ICC) treated with hypofractionated-RT (HF-RT). METHODS Retrospective analysis of 96 patients with unresectable ICC who underwent HF-RT (median 58.05 Gy in 15 fractions) between 2009 and 2022 was performed. Absolute lymphocyte count (ALC) nadir within 12 weeks of RT was analyzed. Primary variable of interest was severe lymphopenia, defined as Grade 3+ (ALC <0.5 k/μL) per CTCAE v5.0. Primary outcome of interest was overall survival (OS) from RT. RESULTS Median follow-up was 16 months. Fifty-two percent of patients had chemotherapy pre-RT, 23% during RT, and 40% post-RT. Pre-RT, median ALC was 1.1 k/μL and 5% had severe lymphopenia. Post-RT, 68% developed RT-related severe lymphopenia. Patients who developed severe lymphopenia had a significantly lower pre-RT ALC (median 1.1 vs. 1.5 k/μL, P =0.01) and larger target tumor volume (median 125 vs. 62 cm 3 , P =0.02). In our multivariable Cox model, severe lymphopenia was associated with a 1.7-fold increased risk of death ( P =0.04); 1-year OS rates were 63% vs 77% ( P =0.03). Receipt of photon versus proton-based RT (OR=3.50, P =0.02), higher mean liver dose (OR=1.19, P <0.01), and longer RT duration (OR=1.49, P =0.02) predicted severe lymphopenia. CONCLUSIONS HF-RT-related lymphopenia is an independent prognostic factor for survival in patients with unresectable ICC. Patients with lower baseline ALC and larger tumor volume may be at increased risk, and use of proton therapy, minimizing mean liver dose, and avoiding treatment breaks may reduce RT-related lymphopenia.
Collapse
Affiliation(s)
- Grace Lee
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Daniel W Kim
- Inova Mather Proton Centre, Inova Schar Cancer Institute, VA
| | - Alicia C Smart
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Nora K Horick
- Massachusetts General Hospital Biostatistics Center, Boston, MA
| | - Christine E Eyler
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC
| | - Hannah J Roberts
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Priyadarshini Pathak
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Lipika Goyal
- Division of Oncology, Department of Medicine, Stanford University Medical Center, Stanford, CA
| | - Joseph Franses
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
| | - James M Heather
- Massachusetts General Hospital Cancer Center and Harvard Medical School Department of Medicine, Boston, MA
| | - William L Hwang
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | | | - Samuel J Klempner
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Lorraine C Drapek
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Jill N Allen
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Lawrence S Blaszkowsky
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Aparna R Parikh
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - David P Ryan
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Jeffrey W Clark
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Jennifer Y Wo
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
18
|
Visa MA, Abazeed ME, Avella Patino D. Integrative Approaches in Non-Small Cell Lung Cancer Management: The Role of Radiotherapy. J Clin Med 2024; 13:4296. [PMID: 39124563 PMCID: PMC11312949 DOI: 10.3390/jcm13154296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Treatment guidelines for non-small cell lung cancer (NSCLC) vary by several factors including pathological stage, patient candidacy, and goal of treatment. With many therapeutics and even more combinations available in the NSCLC clinician's toolkit, a multitude of questions remain unanswered vis-a-vis treatment optimization. While some studies have begun exploring the interplay among the many pillars of NSCLC treatment-surgical resection, radiotherapy, chemotherapy, and immunotherapy-the vast number of combinations and permutations of different therapy modalities in addition to the modulation of each constituent therapy leaves much to be desired in a field that is otherwise rapidly evolving. Given NSCLC's high incidence and lethality, the experimentation of synergistic benefits that combinatorial treatment may confer presents a ripe target for advancement and increased understanding without the cost and burden of novel drug development. This review introduces, synthesizes, and compares prominent NSCLC therapies, placing emphasis on the interplay among types of therapies and the synergistic benefits some combinatorial therapies have demonstrated over the past several years.
Collapse
Affiliation(s)
- Maxime A. Visa
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Mohamed E. Abazeed
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Diego Avella Patino
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| |
Collapse
|
19
|
Sue M, Takeuchi Y, Hirata S, Takaki A, Otsuka M. Impact of Nutritional Status on Neutrophil-to-Lymphocyte Ratio as a Predictor of Efficacy and Adverse Events of Immune Check-Point Inhibitors. Cancers (Basel) 2024; 16:1811. [PMID: 38791890 PMCID: PMC11120021 DOI: 10.3390/cancers16101811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/25/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
The neutrophil -to-lymphocyte ratio (NLR) is useful for predicting the effectiveness of treatment with immune checkpoint inhibitors (ICIs) and immune-related adverse events (irAEs). Because a growing body of evidence has recently shown that the number of lymphocytes that comprise NLR fluctuates according to nutritional status, this study examined whether the usefulness of NLR varies in ICI treatment due to changes in nutritional status. A retrospective analysis was performed on 1234 patients who received ICI treatment for malignant tumors at our hospital. Progression-free survival (PFS) was significantly prolonged in patients with NLR < 4. Multivariate analysis revealed that the factors associated with the occurrence of irAE were NLR < 4 and the use of ipilimumab. However, when limited to cases with serum albumin levels <3.8 g/dL, lymphocyte counts significantly decreased, and the associations between NLR and PFS and between NLR and irAE occurrence disappeared. In contrast, when limited to the cases with serum albumin levels ≥3.8 g/dL, the associations remained, with significantly prolonged PFS and significantly increased irAE occurrence at NLR < 4. NLR may be a good predictive tool for PFS and irAE occurrence during ICI treatment when a good nutritional status is maintained.
Collapse
Affiliation(s)
- Masahiko Sue
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan;
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan; (S.H.); (A.T.); (M.O.)
| | - Yasuto Takeuchi
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan; (S.H.); (A.T.); (M.O.)
- Department of Regenerative Medicine, Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Shoichiro Hirata
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan; (S.H.); (A.T.); (M.O.)
| | - Akinobu Takaki
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan; (S.H.); (A.T.); (M.O.)
| | - Motoyuki Otsuka
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan; (S.H.); (A.T.); (M.O.)
| |
Collapse
|
20
|
Bracamonte-Baran W, Kim ST. The Current and Future of Biomarkers of Immune Related Adverse Events. Rheum Dis Clin North Am 2024; 50:201-227. [PMID: 38670721 PMCID: PMC11232920 DOI: 10.1016/j.rdc.2024.01.004] [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] [Indexed: 04/28/2024]
Abstract
With their groundbreaking clinical responses, immune checkpoint inhibitors (ICIs) have ushered in a new chapter in cancer therapeutics. However, they are often associated with life-threatening or organ-threatening autoimmune/autoinflammatory phenomena, collectively termed immune-related adverse events (irAEs). In this review, we will first describe the mechanisms of action of ICIs as well as irAEs. Next, we will review biomarkers for predicting the development of irAEs or stratifying risks.
Collapse
Affiliation(s)
- William Bracamonte-Baran
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA
| | - Sang T Kim
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA.
| |
Collapse
|
21
|
Zhang GY, Du XZ, Xu R, Chen T, Wu Y, Wu XJ, Liu S. Development and Validation of a Machine Learning-Based Model Using CT Radiomics for Predicting Immune Checkpoint Inhibitor-related Pneumonitis in Patients With NSCLC Receiving Anti-PD1 Immunotherapy: A Multicenter Retrospective CaseControl Study. Acad Radiol 2024; 31:2128-2143. [PMID: 37977890 DOI: 10.1016/j.acra.2023.10.039] [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: 08/24/2023] [Revised: 10/06/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
RATIONALE AND OBJECTIVES This study aimed to develop and evaluate a radiomics-based model combined with clinical and qualitative radiological (semantic feature [SF]) features to predict immune checkpoint inhibitor-related pneumonitis (CIP) in patients with non-small cell lung cancer (NSCLC) treated with programmed cell death protein 1 inhibitors. MATERIALS AND METHODS This was a multicenter retrospective casecontrol study conducted from January 1, 2018, to December 31, 2022, at three centers. Patients with NSCLC treated with anti-PD1 were enrolled and randomly divided into two groups (7:3): training (n = 95) and validation (n = 39). Logistic regression (LR) and support vector machine (SVM) algorithms were used to transform features into the models. RESULTS The study comprised 134 participants from three independent centers (male, 114/134, 85%; mean [±standard deviation] age, 63.92 [±7.9] years). The radiomics score (RS) models built based on the LR and SVM algorithms could accurately predict CIP (area under the receiver operating characteristics curve [AUC], 0.860 [0.780, 0.939] and 0.861 [0.781, 0.941], respectively). The AUCs for the RS-clinic-SF combined model were 0.903 (0.839, 0.967) and 0.826 (0.688, 0.964) in the training and validation cohorts, respectively. Decision curve analysis showed that the combined models achieved high clinical net benefit across the majority of the range of reasonable threshold probabilities. CONCLUSION This study demonstrated that the combined model constructed by the identified features of RS, clinical features, and SF has the potential to precisely predict CIP. The RS-clinic-SF combined model has the potential to be used more widely as a practical tool for the noninvasive prediction of CIP to support individualized treatment planning.
Collapse
Affiliation(s)
- Guo-Yue Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (G.-y.Z., X.-z.D., R.X., Y.W., X.-j.W.).
| | - Xian-Zhi Du
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (G.-y.Z., X.-z.D., R.X., Y.W., X.-j.W.).
| | - Rui Xu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (G.-y.Z., X.-z.D., R.X., Y.W., X.-j.W.).
| | - Ting Chen
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (T.C.).
| | - Yue Wu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (G.-y.Z., X.-z.D., R.X., Y.W., X.-j.W.).
| | - Xiao-Juan Wu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (G.-y.Z., X.-z.D., R.X., Y.W., X.-j.W.); Department of Respiratory and Critical Care Medicine, Suining Central Hospital, Suining, 629000, Sichuan, P.R. China (X.-j.W.).
| | - Shui Liu
- Department of Respiratory and Critical Care Medicine, People's Hospital of Fengjie, Fengjie, Chongqing, 404600, P.R. China (S.L.).
| |
Collapse
|
22
|
Conroy MR, O'Sullivan H, Collins DC, Bambury RM, Power D, Grossman S, O'Reilly S. Exploring the prognostic impact of absolute lymphocyte count in patients treated with immune-checkpoint inhibitors. BJC REPORTS 2024; 2:31. [PMID: 39516713 PMCID: PMC11523911 DOI: 10.1038/s44276-024-00058-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 03/05/2024] [Accepted: 03/17/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The role of immune checkpoint inhibitors (ICI) expands but affordable and reproducible prognostic biomarkers are needed. We investigated the association between baseline and 3-month absolute lymphocyte count (ALC) and survival for patients on ICI. METHODS A retrospective study investigated patients who received ICI July 2014-August 2019. Survival probabilities were calculated for lymphocyte subsets. Univariate and multivariate analyses were performed to investigate risk factors for lymphopenia. RESULTS Among 179 patients, median age was 62 and 41% were female. The most common diagnoses were melanoma (41%) and lung cancer (40%). Median PFS was 6.5 months. 27% had baseline lymphopenia (ALC < 1 × 109cells/L) and no significant difference in PFS or OS to those with normal ALC. However, 31% had lymphopenia at 3 months and significantly shorter OS than those without (9.8 vs 18.3 months, p < 0.001). Those with baseline lymphopenia who recovered counts at 3 months had no difference in PFS (median NR vs 13.0 months, p = 0.48) or OS (22 vs 18.3 months, p = 0.548) to those never lymphopenic. The strongest risk factor for lymphopenia on multivariable analysis was previous radiation therapy (RT). CONCLUSIONS 3-month lymphopenia is a negative prognostic marker in cancer patients on ICI. Previous RT is significantly associated with lymphopenia.
Collapse
Affiliation(s)
- M R Conroy
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
- Cancer research @UCC, Western Gateway Building, Western Road, Cork, Ireland
| | - H O'Sullivan
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
- Cancer research @UCC, Western Gateway Building, Western Road, Cork, Ireland
| | - D C Collins
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
- Cancer research @UCC, Western Gateway Building, Western Road, Cork, Ireland
| | - R M Bambury
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
- Cancer research @UCC, Western Gateway Building, Western Road, Cork, Ireland
| | - D Power
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
- Cancer research @UCC, Western Gateway Building, Western Road, Cork, Ireland
- Mercy University Hospital, Grenville Pl, Centre, Cork, Ireland
| | - S Grossman
- Johns Hopkins University, Baltimore, MD, USA
| | - S O'Reilly
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland.
- Cancer research @UCC, Western Gateway Building, Western Road, Cork, Ireland.
- Mercy University Hospital, Grenville Pl, Centre, Cork, Ireland.
| |
Collapse
|
23
|
Kawano M, Yano Y, Yamamoto A, Yasutomi E, Inoue Y, Kitadai J, Yoshida R, Matsuura T, Shiomi Y, Ueda Y, Kodama Y. Risk Factors for Immune Checkpoint Inhibitor-Induced Liver Injury and the Significance of Liver Biopsy. Diagnostics (Basel) 2024; 14:815. [PMID: 38667461 PMCID: PMC11049019 DOI: 10.3390/diagnostics14080815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/07/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Immune checkpoint inhibitor (ICI)-induced liver injury (LI) is a common adverse event, but the clinical characteristics based on the classification of hepatocellular injury and cholestatic types are not fully evaluated. This study aims to analyze risk factors and histological findings in relation to the classification of ICI-induced LI. In total, 254 ICI-induced LI patients among 1086 treated with ICIs between September 2014 and March 2022 were classified according to the diagnostic criteria for drug-induced LI (DILI), and their risk factors and outcomes were evaluated. Kaplan-Meier analyses showed that overall survival in patients with hepatocellular-injury-type LI was significantly longer than others (p < 0.05). Regarding pre-treatment factors, the lymphocyte count was significantly higher in patients with ICI-induced LI, especially in hepatocellular-injury-type LI. Gamma glutamyl transferase (γGTP) and alkaline phosphatase (ALP) were also significantly lower in patients with ICI-induced LI (p < 0.05). Multivariate analyses revealed that malignant melanoma, high lymphocyte count, and low ALP levels were extracted as factors contributing to hepatocellular-injury-type LI. The histological findings among 37 patients diagnosed as ICI-induced LI via liver biopsy also revealed that the spotty/focal necrosis was significantly frequent in hepatocellular-injury-type LI, whereas ductular reactions were frequently observed in cholestatic-type LI. It is suggested that the histological inflammation pattern in patients with LI is closely correlated with the type of DILI.
Collapse
Affiliation(s)
- Miki Kawano
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.K.); (A.Y.); (Y.I.); (J.K.); (R.Y.); (T.M.); (Y.S.); (Y.U.); (Y.K.)
| | - Yoshihiko Yano
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.K.); (A.Y.); (Y.I.); (J.K.); (R.Y.); (T.M.); (Y.S.); (Y.U.); (Y.K.)
| | - Atsushi Yamamoto
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.K.); (A.Y.); (Y.I.); (J.K.); (R.Y.); (T.M.); (Y.S.); (Y.U.); (Y.K.)
| | - Eiichiro Yasutomi
- Department of Gastroenterology, Hyogo Prefectural Kakogawa Medical Center, Kakogawa 675-8555, Japan;
| | - Yuta Inoue
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.K.); (A.Y.); (Y.I.); (J.K.); (R.Y.); (T.M.); (Y.S.); (Y.U.); (Y.K.)
| | - Jun Kitadai
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.K.); (A.Y.); (Y.I.); (J.K.); (R.Y.); (T.M.); (Y.S.); (Y.U.); (Y.K.)
| | - Ryutaro Yoshida
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.K.); (A.Y.); (Y.I.); (J.K.); (R.Y.); (T.M.); (Y.S.); (Y.U.); (Y.K.)
| | - Takanori Matsuura
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.K.); (A.Y.); (Y.I.); (J.K.); (R.Y.); (T.M.); (Y.S.); (Y.U.); (Y.K.)
| | - Yuuki Shiomi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.K.); (A.Y.); (Y.I.); (J.K.); (R.Y.); (T.M.); (Y.S.); (Y.U.); (Y.K.)
| | - Yoshihide Ueda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.K.); (A.Y.); (Y.I.); (J.K.); (R.Y.); (T.M.); (Y.S.); (Y.U.); (Y.K.)
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (M.K.); (A.Y.); (Y.I.); (J.K.); (R.Y.); (T.M.); (Y.S.); (Y.U.); (Y.K.)
| |
Collapse
|
24
|
Jani Y, Jansen CS, Gerke MB, Bilen MA. Established and emerging biomarkers of immunotherapy in renal cell carcinoma. Immunotherapy 2024; 16:405-426. [PMID: 38264827 PMCID: PMC11913054 DOI: 10.2217/imt-2023-0267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/08/2024] [Indexed: 01/25/2024] Open
Abstract
Immunotherapies, such as immune checkpoint inhibitors, have heralded impressive progress for patient care in renal cell carcinoma (RCC). Despite this success, some patients' disease fails to respond, and other patients experience significant side effects. Thus, development of biomarkers is needed to ensure that patients can be selected to maximize benefit from immunotherapies. Improving clinicians' ability to predict which patients will respond to immunotherapy and which are most at risk of adverse events - namely through clinical biomarkers - is indispensable for patient safety and therapeutic efficacy. Accordingly, an evolving suite of therapeutic biomarkers continues to be investigated. This review discusses biomarkers for immunotherapy in RCC, highlighting current practices and emerging innovations, aiming to contribute to improved outcomes for patients with RCC.
Collapse
Affiliation(s)
- Yash Jani
- Mercer University, Macon, GA31207, USA
| | - Caroline S Jansen
- Emory University School of Medicine, Atlanta, GA30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA30322, USA
| | - Margo B Gerke
- Emory University School of Medicine, Atlanta, GA30322, USA
| | - Mehmet Asim Bilen
- Winship Cancer Institute of Emory University, Atlanta, GA30322, USA
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Atlanta, GA30322, USA
| |
Collapse
|
25
|
Tokunaga T, Aoki M, Maruyama K, Nonaka Y, Kariatsumari K, Sakasegawa K, Ueda K. Fulminant myocarditis during postoperative adjuvant chemotherapy for lung cancer with atezolizumab: a case report. J Med Case Rep 2024; 18:162. [PMID: 38491548 PMCID: PMC10943901 DOI: 10.1186/s13256-024-04447-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/09/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Postoperative adjuvant systemic therapy with atezolizumab for lung cancer has been reported to be effective. Although myocarditis is a rare immune adverse event associated with atezolizumab, it can have a serious course and should be treated with caution. We herein report a case of fulminant myocarditis during adjuvant systemic therapy with atezolizumab. CASE PRESENTATION The patient was a 49-year-old Asian woman. She was diagnosed with pT2aN1M0 stage IIB (Programmed Death Ligand 1(PD-L1), 50%) after surgery for right upper lobe lung adenocarcinoma. Atezolizumab was administered following platinum-based adjuvant chemotherapy. On day 14, the patient was hospitalized because of deterioration in her general condition caused by fever. On day 16, she developed dyspnea, which worsened, and on day 17, she experienced shock. Blood tests, echocardiography, and cardiac catheterization were performed, and the patient was diagnosed with cardiogenic shock due to myocarditis. Initial measures did not improve the patient's shock state. The patient was transferred to hospital for the use of an assistive circulatory system. Pulse steroid therapy was administered, and myocarditis showed a tendency toward improvement. A retrospective review of the patient's history revealed a decreased lymphocyte count and an increase in the neutrophil/lymphocyte ratio, which may be useful for detecting severe immune-related adverse events. The troponin levels were elevated, but creatine phosphokinase level remained within the normal range. CONCLUSION Myocarditis can be fatal due to the rapid progression of symptoms. Close follow-up, a prompt diagnosis, and therapeutic intervention are important. Decreased lymphocyte counts, increased neutrophil/lymphocyte ratios, and the measurement of multiple myocardial biomarkers are considered useful for the early diagnosis of myocarditis.
Collapse
Affiliation(s)
- Takuya Tokunaga
- Department of General Thoracic Surgery, Kagoshima City Hospital, 37-1, Uearatacho, Kagoshima, 890-8760, Japan.
- Department of General Thoracic Surgery, Kagoshima University Graduate School of Dental and Medical Science, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan.
| | - Masaya Aoki
- Department of General Thoracic Surgery, Kagoshima University Graduate School of Dental and Medical Science, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Koki Maruyama
- Department of General Thoracic Surgery, Kagoshima City Hospital, 37-1, Uearatacho, Kagoshima, 890-8760, Japan
| | - Yuto Nonaka
- Department of General Thoracic Surgery, Kagoshima City Hospital, 37-1, Uearatacho, Kagoshima, 890-8760, Japan
| | - Kota Kariatsumari
- Department of General Thoracic Surgery, Kagoshima City Hospital, 37-1, Uearatacho, Kagoshima, 890-8760, Japan
| | - Koichi Sakasegawa
- Department of General Thoracic Surgery, Kagoshima City Hospital, 37-1, Uearatacho, Kagoshima, 890-8760, Japan
| | - Kazuhiro Ueda
- Department of General Thoracic Surgery, Kagoshima University Graduate School of Dental and Medical Science, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| |
Collapse
|
26
|
Lin MX, Zang D, Liu CG, Han X, Chen J. Immune checkpoint inhibitor-related pneumonitis: research advances in prediction and management. Front Immunol 2024; 15:1266850. [PMID: 38426102 PMCID: PMC10902117 DOI: 10.3389/fimmu.2024.1266850] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
The advent of immune-checkpoint inhibitors (ICIs) has revolutionized the treatment of malignant solid tumors in the last decade, producing lasting benefits in a subset of patients. However, unattended excessive immune responses may lead to immune-related adverse events (irAEs). IrAEs can manifest in different organs within the body, with pulmonary toxicity commonly referred to as immune checkpoint inhibitor-related pneumonitis (CIP). The CIP incidence remains high and is anticipated to rise further as the therapeutic indications for ICIs expand to encompass a wider range of malignancies. The diagnosis and treatment of CIP is difficult due to the large individual differences in its pathogenesis and severity, and severe CIP often leads to a poor prognosis for patients. This review summarizes the current state of clinical research on the incidence, risk factors, predictive biomarkers, diagnosis, and treatment for CIP, and we address future directions for the prevention and accurate prediction of CIP.
Collapse
Affiliation(s)
| | | | | | | | - Jun Chen
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
27
|
Stepanenko AA, Sosnovtseva AO, Valikhov MP, Chernysheva AA, Abramova OV, Naumenko VA, Chekhonin VP. The need for paradigm shift: prognostic significance and implications of standard therapy-related systemic immunosuppression in glioblastoma for immunotherapy and oncolytic virotherapy. Front Immunol 2024; 15:1326757. [PMID: 38390330 PMCID: PMC10881776 DOI: 10.3389/fimmu.2024.1326757] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Despite significant advances in our knowledge regarding the genetics and molecular biology of gliomas over the past two decades and hundreds of clinical trials, no effective therapeutic approach has been identified for adult patients with newly diagnosed glioblastoma, and overall survival remains dismal. Great hopes are now placed on combination immunotherapy. In clinical trials, immunotherapeutics are generally tested after standard therapy (radiation, temozolomide, and steroid dexamethasone) or concurrently with temozolomide and/or steroids. Only a minor subset of patients with progressive/recurrent glioblastoma have benefited from immunotherapies. In this review, we comprehensively discuss standard therapy-related systemic immunosuppression and lymphopenia, their prognostic significance, and the implications for immunotherapy/oncolytic virotherapy. The effectiveness of immunotherapy and oncolytic virotherapy (viro-immunotherapy) critically depends on the activity of the host immune cells. The absolute counts, ratios, and functional states of different circulating and tumor-infiltrating immune cell subsets determine the net immune fitness of patients with cancer and may have various effects on tumor progression, therapeutic response, and survival outcomes. Although different immunosuppressive mechanisms operate in patients with glioblastoma/gliomas at presentation, the immunological competence of patients may be significantly compromised by standard therapy, exacerbating tumor-related systemic immunosuppression. Standard therapy affects diverse immune cell subsets, including dendritic, CD4+, CD8+, natural killer (NK), NKT, macrophage, neutrophil, and myeloid-derived suppressor cell (MDSC). Systemic immunosuppression and lymphopenia limit the immune system's ability to target glioblastoma. Changes in the standard therapy are required to increase the success of immunotherapies. Steroid use, high neutrophil-to-lymphocyte ratio (NLR), and low post-treatment total lymphocyte count (TLC) are significant prognostic factors for shorter survival in patients with glioblastoma in retrospective studies; however, these clinically relevant variables are rarely reported and correlated with response and survival in immunotherapy studies (e.g., immune checkpoint inhibitors, vaccines, and oncolytic viruses). Our analysis should help in the development of a more rational clinical trial design and decision-making regarding the treatment to potentially improve the efficacy of immunotherapy or oncolytic virotherapy.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasiia O. Sosnovtseva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marat P. Valikhov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasia A. Chernysheva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga V. Abramova
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Victor A. Naumenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
28
|
Kovacsovics-Bankowski M, Sweere JM, Healy CP, Sigal N, Cheng LC, Chronister WD, Evans SA, Marsiglio J, Gibson B, Swami U, Erickson-Wayman A, McPherson JP, Derose YS, Eliason AL, Medina CO, Srinivasan R, Spitzer MH, Nguyen N, Hyngstrom J, Hu-Lieskovan S. Lower frequencies of circulating suppressive regulatory T cells and higher frequencies of CD4 + naïve T cells at baseline are associated with severe immune-related adverse events in immune checkpoint inhibitor-treated melanoma. J Immunother Cancer 2024; 12:e008056. [PMID: 38233101 PMCID: PMC10806651 DOI: 10.1136/jitc-2023-008056] [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: 12/18/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Immune-related adverse events (irAEs) are major barriers of clinical management and further development of immune checkpoint inhibitors (ICIs) for cancer therapy. Therefore, biomarkers associated with the onset of severe irAEs are needed. In this study, we aimed to identify immune features detectable in peripheral blood and associated with the development of severe irAEs that required clinical intervention. METHODS We used a 43-marker mass cytometry panel to characterize peripheral blood mononuclear cells from 28 unique patients with melanoma across 29 lines of ICI therapy before treatment (baseline), before the onset of irAEs (pre-irAE) and at the peak of irAEs (irAE-max). In the 29 lines of ICI therapy, 18 resulted in severe irAEs and 11 did not. RESULTS Unsupervised and gated population analysis showed that patients with severe irAEs had a higher frequency of CD4+ naïve T cells and lower frequency of CD16+ natural killer (NK) cells at all time points. Gated population analysis additionally showed that patients with severe irAEs had fewer T cell immunoreceptor with Ig and ITIM domain (TIGIT+) regulatory T cells at baseline and more activated CD38+ CD4+ central memory T cells (TCM) and CD39+ and Human Leukocyte Antigen-DR Isotype (HLA-DR)+ CD8+ TCM at peak of irAEs. The differentiating immune features at baseline were predominantly seen in patients with gastrointestinal and cutaneous irAEs and type 1 diabetes. Higher frequencies of CD4+ naïve T cells and lower frequencies of CD16+ NK cells were also associated with clinical benefit to ICI therapy. CONCLUSIONS This study demonstrates that high-dimensional immune profiling can reveal novel blood-based immune signatures associated with risk and mechanism of severe irAEs. Development of severe irAEs in melanoma could be the result of reduced immune inhibitory capacity pre-ICI treatment, resulting in more activated TCM cells after treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - John Marsiglio
- The University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Berit Gibson
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Umang Swami
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Alyssa Erickson-Wayman
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jordan P McPherson
- Department of Pharmacy, Huntsman Cancer Institute Cancer Hospital, Salt Lake City, Utah, USA
| | - Yoko S Derose
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | | | | | | | - Matthew H Spitzer
- Teiko.bio Inc, Salt Lake City, Utah, USA
- Department of Otolaryngology-Head and Neck Cancer, University of California San Francisco, San Francisco, California, USA
| | | | - John Hyngstrom
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Siwen Hu-Lieskovan
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
29
|
Li Y, Pond G, McWhirter E. Multisystem Immune-Related Adverse Events from Dual-Agent Immunotherapy Use. Curr Oncol 2024; 31:425-435. [PMID: 38248113 PMCID: PMC10813982 DOI: 10.3390/curroncol31010028] [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: 10/17/2023] [Revised: 11/27/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND little is known about the incidence and characteristics of multisystem immune-related adverse events (irAEs) associated with dual-agent ipilimumab and nivolumab use. METHODS A retrospective cohort review was completed that included cancer patients seen at the Juravinski Cancer Centre who received at least one dose of ipilimumab and nivolumab from 2018 to 2022. Patient characteristics, cancer types, and irAEs were recorded. Multivariate logistic and cox regressions were completed, comparing those who developed multisystem irAEs, single irAE, and no irAE. RESULTS A total of 123 patients were included in this study. Out of 123 patients, 72 (59%) had melanoma, 50/123 (41%) had renal cell carcinoma (RCC), and 1/123 (1%) had breast cancer. Multisystem irAEs were seen in 40% of the overall cohort. The most common irAE type was dermatitis (22%), followed by colitis (19%) and hepatitis (17%). CONCLUSIONS Our study demonstrated that multisystem irAEs are prevalent amongst patients receiving ipilimumab and nivolumab. It is important for both physician education and the counseling and consent of patients to monitor the potential for multiple irAEs.
Collapse
Affiliation(s)
- Yuchen Li
- Department of Oncology, McMaster University, Hamilton, ON L8V 5C2, Canada
| | - Gregory Pond
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Elaine McWhirter
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| |
Collapse
|
30
|
Galts A, Hammi A. FLASH radiotherapy sparing effect on the circulating lymphocytes in pencil beam scanning proton therapy: impact of hypofractionation and dose rate. Phys Med Biol 2024; 69:025006. [PMID: 38081067 DOI: 10.1088/1361-6560/ad144e] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/11/2023] [Indexed: 01/06/2024]
Abstract
Purpose. The sparing effect of ultra-high dose rate (FLASH) radiotherapy has been reported, but its potential to mitigate depletion of circulating blood and lymphocytes (CL) has not been investigated in pencil-beam scanning-based (PBS) proton therapy, which could potentially reduce the risk of radiation-induced lymphopenia.Material and methods. A time-dependent framework was used to score the dose to the CL during the course of radiotherapy. For brain patients, cerebral vasculatures were semi-automatic segmented from 3T MR-angiography data. A dynamic beam delivery system was developed capable of simulating spatially varying instantaneous dose rates of PBS treatment plans, and which is based on realistic beam delivery parameters that are available clinically. We simulated single and different hypofractionated PBS intensity modulated proton therapy (IMPT) FLASH schemes using 600 nA beam current along with conventionally fractionated IMPT treatment plan at 2 nA beam current. The dosimetric impact of treatment schemes on CL was quantified, and we also evaluated the depletion in subsets of CL based on their radiosensitivity.Results. The proton FLASH sparing effect on CL was observed. In single-fraction PBS FLASH, just 1.5% of peripheral blood was irradiated, whereas hypofractionated FLASH irradiated 7.3% of peripheral blood. In contrast, conventional fractionated IMPT exposed 42.4% of peripheral blood to radiation. PBS FLASH reduced the depletion rate of CL by 69.2% when compared to conventional fractionated IMPT.Conclusion. Our dosimetric blood flow model provides quantitative measures of the PBS FLASH sparing effect on the CL in radiotherapy for brain cancer. FLASH Single treatment fraction offers superior CL sparing when compared to hypofractionated FLASH and conventional IMPT, supporting assumptions about reducing risks of lymphopenia compared to proton therapy at conventional dose rates. The results also indicate that faster conformal FLASH delivery, such as passive patient-specific energy modulation, may further enhance the sparing of the immune system.
Collapse
|
31
|
Kuwano A, Yada M, Tanaka K, Koga Y, Nagasawa S, Masumoto A, Motomura K. Neutrophil-to-Lymphocyte Ratio Predicts Immune-related Adverse Events in Patients With Hepatocellular Carcinoma Treated With Atezolizumab Plus Bevacizumab. CANCER DIAGNOSIS & PROGNOSIS 2024; 4:34-41. [PMID: 38173658 PMCID: PMC10758843 DOI: 10.21873/cdp.10282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/06/2023] [Indexed: 01/05/2024]
Abstract
Background/Aim Atezolizumab in combination with bevacizumab is an approved systemic chemotherapy regimen for advanced hepatocellular carcinoma (HCC). However, immune checkpoint inhibitors (ICIs), such as atezolizumab, frequently lead to immune-related adverse events (irAEs). The identification of biomarkers that can predict the occurrence of irAEs is crucial for the optimal management of patients undergoing ICI treatment. Patients and Methods Between October 2020 and June 2023, we conducted a study involving 69 patients with advanced HCC who received treatment with atezolizumab plus bevacizumab. We conducted an analysis of blood-based biomarkers to identify independent risk factors associated with irAEs. Results In our study, 12 out of 69 patients (17.4%) experienced irAEs. Our investigation into blood-based biomarkers revealed that a neutrophil-to-lymphocyte ratio (NLR) <2.04 at three weeks after the initiation of treatment had high predictive power (area under the curve: 0.77) for irAEs. Furthermore, multivariate logistic analysis identified NLR at three weeks (hazard ratio=0.23; p=0.037) and non-viral infection (hazard ratio=4.47; p=0.037) as independent factors contributing to the occurrence of irAEs. Patients who developed irAEs demonstrated a more favorable overall response rate (75.0% vs. 28.1%, p=0.005), disease control rate (91.6% vs. 52.6%, p=0.016), and progression-free survival (12.1 months vs. 6.0 months, p=0.010) than those who did not experience irAEs. Conclusion An NLR <2.04 at three weeks after the initiation of treatment may serve as a valuable biomarker for predicting irAEs in patients with HCC undergoing atezolizumab plus bevacizumab therapy.
Collapse
Affiliation(s)
- Akifumi Kuwano
- Department of Hepatology, Aso Iizuka Hospital, Fukuoka, Japan
| | - Masayoshi Yada
- Department of Hepatology, Aso Iizuka Hospital, Fukuoka, Japan
| | - Kosuke Tanaka
- Department of Hepatology, Aso Iizuka Hospital, Fukuoka, Japan
| | - Yuta Koga
- Department of Hepatology, Aso Iizuka Hospital, Fukuoka, Japan
| | | | | | - Kenta Motomura
- Department of Hepatology, Aso Iizuka Hospital, Fukuoka, Japan
| |
Collapse
|
32
|
Teijeira L, Martínez M, Moreno A, de Elejoste I, Ibáñez-Beroiz B, Arrazubi V, Díaz de Corcuera I, Elejalde I, Campillo-Calatayud A, Les I. Baseline Circulating Blood Cell Counts and Ratios and Changes Therein for Predicting Immune-Related Adverse Events during Immune Checkpoint Inhibitor Therapy: A Multicenter, Prospective, Observational, Pan-Cancer Cohort Study with a Gender Perspective. Cancers (Basel) 2023; 16:151. [PMID: 38201577 PMCID: PMC10778233 DOI: 10.3390/cancers16010151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 12/21/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Several factors have been associated with the occurrence of immune-related adverse events (irAEs) induced by immune checkpoint inhibitor (ICI) therapy. Despite their availability, the predictive value of circulating blood cell parameters remains underexplored. Our aim was to investigate whether baseline values of and early changes in absolute neutrophil count (ANC), absolute lymphocyte count (ALC), other blood cell counts, and lymphocyte-related ratios can predict irAEs and whether sex may differentially influence this potential predictive ability. Of the 145 patients included, 52 patients (35.8%) experienced at least one irAE, with a 1-year cumulative incidence of 41.6%. Using Fine and Gray competing risk models, we identified female sex (hazard ratio (HR) = 2.17, 95% confidence interval (CI) = 1.20-3.85), high ALC before ICI initiation (HR = 1.63, 95% CI = 1.09-2.45), and low ANC after ICI initiation (HR = 0.81, 95% CI = 0.69-0.96) as predictors of irAEs. However, ALC and ANC may only have an impact on the risk of irAEs in women (stratified for female sex, ALC-related HR = 2.61, 95% CI = 1.40-4.86 and ANC-related HR = 0.57, 95% CI = 0.41-0.81). Priority should be given to developing models to predict ICI-related toxicity and their validation in various settings, and such models should assess the impact of patient sex on the risk of toxicity.
Collapse
Affiliation(s)
- Lucía Teijeira
- Servicio de Oncología Médica, Hospital Universitario de Navarra, 31008 Pamplona, Spain; (L.T.); (V.A.)
| | - Mireia Martínez
- Servicio de Oncología Médica, Hospital Universitario Araba, Servicio Vasco de Salud Osakidetza, 01009 Vitoria-Gasteiz, Spain;
- Grupo de Investigación en Cáncer de Pulmón, Instituto de Investigación Sanitaria Bioaraba, 01009 Vitoria-Gasteiz, Spain
| | - Amaia Moreno
- Servicio de Oncología Médica, Hospital Universitario Galdakao, 48960 Galdácano, Spain; (A.M.); (I.D.d.C.)
| | - Ibone de Elejoste
- Servicio de Oncología Médica, Hospital Universitario Donostia, 20014 San Sebastián, Spain;
| | - Berta Ibáñez-Beroiz
- Servicio de Metodología, IdISNA, Navarrabiomed-Universidad Pública de Navarra, 31008 Pamplona, Spain;
| | - Virginia Arrazubi
- Servicio de Oncología Médica, Hospital Universitario de Navarra, 31008 Pamplona, Spain; (L.T.); (V.A.)
| | - Isabela Díaz de Corcuera
- Servicio de Oncología Médica, Hospital Universitario Galdakao, 48960 Galdácano, Spain; (A.M.); (I.D.d.C.)
| | - Iñaki Elejalde
- Servicio de Medicina Interna, Hospital Universitario de Navarra, 31008 Pamplona, Spain;
- Unidad de Enfermedades Autoinmunes Sistémicas, Servicio de Medicina Interna, Hospital Universitario de Navarra, 31008 Pamplona, Spain
- Grupo de Enfermedades Inflamatorias e Inmunomediadas, IdISNA, Navarrabiomed-Universidad Pública de Navarra, 31008 Pamplona, Spain;
| | - Ana Campillo-Calatayud
- Grupo de Enfermedades Inflamatorias e Inmunomediadas, IdISNA, Navarrabiomed-Universidad Pública de Navarra, 31008 Pamplona, Spain;
| | - Iñigo Les
- Servicio de Medicina Interna, Hospital Universitario de Navarra, 31008 Pamplona, Spain;
- Unidad de Enfermedades Autoinmunes Sistémicas, Servicio de Medicina Interna, Hospital Universitario de Navarra, 31008 Pamplona, Spain
- Grupo de Enfermedades Inflamatorias e Inmunomediadas, IdISNA, Navarrabiomed-Universidad Pública de Navarra, 31008 Pamplona, Spain;
- Departamento de Ciencias de la Salud, Universidad Pública de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
33
|
Yildirim HC, Kus F, Guven DC, Karaca E, Kaygusuz Y, Dizdar O, Aksoy S, Erman M, Yalcin S, Kilickap S. Mean Platelet Volume to Lymphocyte Ratio: A New Biomarker Predicting Response in Patients with Solid Tumors Treated with Nivolumab. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2023; 6:170-176. [PMID: 38143956 PMCID: PMC10734395 DOI: 10.36401/jipo-23-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/30/2023] [Accepted: 08/15/2023] [Indexed: 12/26/2023]
Abstract
Introduction Although immune checkpoint inhibitors (ICIs) are widely used in cancer treatment, identifying factors that predict treatment response remains a challenge in clinical practice. There is a need for biomarkers to identify patients who may not benefit from these treatments. It is crucial to identify a simple and cost-effective biomarker that can be easily incorporated into clinical practice. This study aims to investigate the mean platelet volume to lymphocyte ratio (MPVLR), as measured by a hemogram test, and median overall survival (mOS) in patients with cancer treated with nivolumab. Methods A total of 131 adult patients with metastatic cancer, including malignant melanoma (MM), renal cell carcinoma (RCC), non-small cell lung cancer (NSCLC), and head and neck cancer (HNC), were included in this study. Baseline demographics, ECOG (Eastern Cooperative Oncology Group) performance status, tumor type, and blood count parameters were recorded. Univariate and multivariate analyses were conducted to evaluate potential risk factors. Results The median age of the patients was 59.87 ± 11.97 years, and the median follow-up period was 20.20 months (IQR, 12.80-27.60). RCC (43.5%) and MM (25.9%) were the most common diagnoses. Patients with ECOG scores of 0-1 had a longer mOS than those with scores of 2-3 (mOS: 20.60 months [95% CI, 14.94-25.29] vs. 5.24 months [95% CI, 0-16.42], p < 0.001). Additionally, patients with lactate dehydrogenase (LDH) levels within the normal range had a longer mOS than those with high LDH levels (mOS: 24.54 months [95% CI, 14.13-34.96] vs. 13.10 months [95% CI, 4.49-21.72], p = 0.038). Patients with low MPVLR also had a longer mOS than those with high MPVLR (mOS: 33.70 months [95% CI, 25.99-41.42] vs. 11.07 months [95% CI, 6.89-15.24], p < 0.001). In the multivariate Cox regression analysis, high MPVLR, ECOG score of 2-3, and high LDH level were associated with shorter mOS (p < 0.001, p = 0.001, and p = 0.046, respectively). Conclusion This study demonstrates that MPVLR could serve as a novel biomarker for predicting response to nivolumab treatment. Incorporating MPVLR into clinical practice may aid in identifying patients who are less likely to benefit from the treatment.
Collapse
Affiliation(s)
- Hasan Cagri Yildirim
- Department of Medical Oncology, Hacettepe University Medical School, Ankara, Türkiye
| | - Fatih Kus
- Department of Medical Oncology, Hacettepe University Medical School, Ankara, Türkiye
| | - Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Medical School, Ankara, Türkiye
| | - Ece Karaca
- Department of Internal Medicine, Hacettepe University Medical School, Ankara, Türkiye
| | - Yunus Kaygusuz
- Department of Internal Medicine, Hacettepe University Medical School, Ankara, Türkiye
| | - Omer Dizdar
- Department of Medical Oncology, Hacettepe University Medical School, Ankara, Türkiye
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Medical School, Ankara, Türkiye
| | - Mustafa Erman
- Department of Medical Oncology, Hacettepe University Medical School, Ankara, Türkiye
| | - Suayib Yalcin
- Department of Medical Oncology, Hacettepe University Medical School, Ankara, Türkiye
| | | |
Collapse
|
34
|
Takimoto R, Kamigaki T, Ito H, Saito M, Takizawa K, Soejima K, Yasuda H, Ohgino K, Terai H, Tomita K, Miura M, Mizukoshi E, Miyashita T, Nakamoto Y, Hayashi K, Miwa S, Kitahara M, Takeuchi A, Kimura H, Mochizuki T, Sugie H, Seino KI, Yamada T, Takeuchi S, Makita K, Naitoh K, Yasumoto K, Yoshida Y, Inoue H, Kotake K, Ohshima K, Noda SE, Okamoto M, Yoshimoto Y, Okada S, Ibe H, Oguma E, Goto S. Safety evaluation of immune-cell therapy for malignant tumor in the Cancer Immune-cell Therapy Evaluation Group (CITEG). Cytotherapy 2023; 25:1229-1235. [PMID: 37486281 DOI: 10.1016/j.jcyt.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/10/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND AIMS With the aim of strengthening the scientific evidence of immune-cell therapy for cancer and further examining its safety, in October 2015, our hospital jointly established the Cancer Immune-Cell Therapy Evaluation Group (CITEG) with 39 medical facilities nationwide. METHODS Medical information, such as patients' background characteristics, clinical efficacy and therapeutic cell types obtained from each facility, has been accumulated, analyzed and evaluated by CITEG. In this prospective study, we analyzed the adverse events associated with immune-cell therapy until the end of September 2022, and we presented our interim safety evaluation. RESULTS A total of 3839 patients with malignant tumor were treated with immune-cell therapy, with a median age of 64 years (range, 13-97 years) and a male-to-female ratio of 1:1.08 (1846:1993). Most patients' performance status was 0 or 1 (86.8%) at the first visit, and 3234 cases (84.2%) were advanced or recurrent cases, which accounted for the majority. The total number of administrations reported in CITEG was 31890, of which 960 (3.0%) showed adverse events. The numbers of adverse events caused by treatment were 363 (1.8%) of 19661 administrations of αβT cell therapy, 9 of 845 administrations of γδT-cell therapy (1.1%) and 10 of 626 administrations of natural killer cell therapy (1.6%). The number of adverse events caused by dendritic cell (DC) vaccine therapy was 578 of 10748 administrations (5.4%), which was significantly larger than those for other treatments. Multivariate analysis revealed that αβT cell therapy had a significantly greater risk of adverse events at performance status 1 or higher, and patients younger than 64 years, women or adjuvant immune-cell therapy had a greater risk of adverse events in DC vaccine therapy. Injection-site reactions were the most frequently reported adverse events, with 449 events, the majority of which were associated with DC vaccine therapy. Among all other adverse events, fever (228 events), fatigue (141 events) and itching (131 events) were frequently reported. In contrast, three patients had adverse events (fever, abdominal pain and interstitial pneumonia) that required hospitalization, although they were weakly related to this therapy; rather, it was considered to be the effect of treatment for the primary disease. CONCLUSIONS Immune-cell therapy for cancer was considered to be a safe treatment without serious adverse events.
Collapse
Affiliation(s)
- Rishu Takimoto
- Seta Clinic Group, Tokyo, Japan; Next Generation Cell and Immunotherapy, Advanced Research Institute for Health Science, Juntendo University, Tokyo, Japan; LSI Sapporo Clinic, Sapporo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan.
| | - Takashi Kamigaki
- Seta Clinic Group, Tokyo, Japan; Next Generation Cell and Immunotherapy, Advanced Research Institute for Health Science, Juntendo University, Tokyo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Hisao Ito
- Seta Clinic Group, Tokyo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Masashi Saito
- Seta Clinic Group, Tokyo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Ken Takizawa
- Seta Clinic Group, Tokyo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Kenzo Soejima
- Seta Clinic Group, Tokyo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Hiroyuki Yasuda
- Seta Clinic Group, Tokyo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Keiko Ohgino
- Seta Clinic Group, Tokyo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Hideki Terai
- Seta Clinic Group, Tokyo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Katsuro Tomita
- Kanazawa Advanced Medical Center, Kanazawa City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Miyabi Miura
- Kanazawa Advanced Medical Center, Kanazawa City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Eishiro Mizukoshi
- Kanazawa Advanced Medical Center, Kanazawa City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Tomoharu Miyashita
- Kanazawa Advanced Medical Center, Kanazawa City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Yasunari Nakamoto
- Kanazawa Advanced Medical Center, Kanazawa City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Katsuhiro Hayashi
- Kanazawa Advanced Medical Center, Kanazawa City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Shinji Miwa
- Kanazawa Advanced Medical Center, Kanazawa City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Masaaki Kitahara
- Kanazawa Advanced Medical Center, Kanazawa City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Akihiko Takeuchi
- Kanazawa Advanced Medical Center, Kanazawa City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Hiroaki Kimura
- Kanazawa Advanced Medical Center, Kanazawa City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Takafumi Mochizuki
- Kanazawa Advanced Medical Center, Kanazawa City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Hiroki Sugie
- LSI Sapporo Clinic, Sapporo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Ken-Ichiro Seino
- LSI Sapporo Clinic, Sapporo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Tomonori Yamada
- LSI Sapporo Clinic, Sapporo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Syuhei Takeuchi
- LSI Sapporo Clinic, Sapporo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Kaori Makita
- Kitaosaka Medical Clinic, Suita City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Keiko Naitoh
- Fukuoka Medical Clinic, Fukuoka City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Kosei Yasumoto
- Fukuoka Medical Clinic, Fukuoka City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Yoichiro Yoshida
- Fukuoka Medical Clinic, Fukuoka City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Hiroyuki Inoue
- Fukuoka Medical Clinic, Fukuoka City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Katsuhiro Kotake
- Masuko Memorial Hospital, Nagoya City, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Kihachi Ohshima
- Heisei-Hidaka Clinic, Gunma, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Shin-Ei Noda
- Heisei-Hidaka Clinic, Gunma, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Masahiko Okamoto
- Heisei-Hidaka Clinic, Gunma, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Yuya Yoshimoto
- Heisei-Hidaka Clinic, Gunma, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| | - Sachiko Okada
- Seta Clinic Group, Tokyo, Japan; Next Generation Cell and Immunotherapy, Advanced Research Institute for Health Science, Juntendo University, Tokyo, Japan
| | - Hiroshi Ibe
- Seta Clinic Group, Tokyo, Japan; Next Generation Cell and Immunotherapy, Advanced Research Institute for Health Science, Juntendo University, Tokyo, Japan
| | - Eri Oguma
- Seta Clinic Group, Tokyo, Japan; Next Generation Cell and Immunotherapy, Advanced Research Institute for Health Science, Juntendo University, Tokyo, Japan
| | - Shigenori Goto
- Seta Clinic Group, Tokyo, Japan; Next Generation Cell and Immunotherapy, Advanced Research Institute for Health Science, Juntendo University, Tokyo, Japan; Cancer Immune-cell Therapy Evaluation Group (CITEG), Tokyo, Japan
| |
Collapse
|
35
|
Koukourakis IM, Giakzidis AG, Koukourakis MI. Anti-PD-1 immunotherapy with dose-adjusted ultra-hypofractionated re-irradiation in patients with locoregionally recurrent head and neck cancer. Clin Transl Oncol 2023; 25:3032-3041. [PMID: 37059932 PMCID: PMC10462536 DOI: 10.1007/s12094-023-03172-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 03/21/2023] [Indexed: 04/16/2023]
Abstract
INTRODUCTION Patients with recurrent inoperable squamous-cell head-neck cancer (HNSCC) after chemo-radiotherapy have an ominous prognosis. Re-irradiation can be applied with some efficacy and high toxicity rates. Anti-PD-1 immunotherapy is effective in 25% of patients. Immunogenic death produced by large radiotherapy (RT) fractions may enhance immune response. MATERIALS AND METHODS We evaluated the efficacy and tolerance of ultra-hypofractionated immuno-radiotherapy (uhypo-IRT) in 17 patients with recurrent HNSCC and 1 with melanoma. Four of HNSCC patients also had oligometastatic disease. Using a dose/time/toxicity-based algorithm, 7, 7 and 4 patients received 1, 2 and 3 fractions of 8 Gy to the tumor, respectively. Nivolumab anti-PD-1 immunotherapy was administered concurrently with RT and continued for 24 cycles, or until disease progression or manifestation of immune-related adverse events (irAEs). RESULTS Early and late RT toxicities were minimal. Three patients developed irAEs (16%). After the 12th cycle, 7/17 (41.2%) and 5/17 (29.4%) patients with HNSCC showed complete (CR) and partial response (PR), respectively. CR was also achieved in the melanoma patient. The objective response rates in HNSCC patients were 57%, 86% and 66%, after 1, 2 and 3 fractions, respectively (overall response rate 70.6%). Most responders experienced an increase in peripheral lymphocyte counts. The median time to progression was 10 months. The 3-year projected locoregional progression-free survival was 35%, while the 3-year disease-specific overall survival was 50%. CONCLUSIONS Anti-PD1 uhypo-IRT is safe and effective in patients with recurrent HNSCC. The high objective response rates and the long survival without evidence of disease support further trials on uhypo-IRT.
Collapse
Affiliation(s)
- Ioannis M Koukourakis
- Radiation Oncology Unit, 1st, Department of Radiology, Medical School, "Aretaieion" University Hospital, National and Kapodistrian University of Athens (NKUOA), Athens, Greece
- Department of Radiotherapy - Oncology, Medical School, Democritus University of Thrace, 68100, Alexandroupolis, Greece
| | - Axiotis G Giakzidis
- Department of Radiotherapy - Oncology, Medical School, Democritus University of Thrace, 68100, Alexandroupolis, Greece
| | - Michael I Koukourakis
- Department of Radiotherapy - Oncology, Medical School, Democritus University of Thrace, 68100, Alexandroupolis, Greece.
| |
Collapse
|
36
|
Tasaki Y, Hamamoto S, Sugiyama Y, Tomiyama N, Naiki T, Etani T, Taguchi K, Matsuyama N, Sue Y, Mimura Y, Kubota H, Noda Y, Aoki M, Moritoki Y, Nozaki S, Kurokawa S, Okada A, Kawai N, Yasui T, Kimura K. Elevated eosinophils proportion as predictor of immune-related adverse events after ipilimumab and nivolumab treatment of advanced and metastatic renal cell carcinoma. Int J Urol 2023; 30:866-874. [PMID: 37278575 DOI: 10.1111/iju.15220] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 05/22/2023] [Indexed: 06/07/2023]
Abstract
OBJECTIVES Ipilimumab and nivolumab treatment against advanced and metastatic renal cell carcinoma (RCC) causes severe and lethal immune-related adverse events (irAEs). Predicting irAEs might improve clinical outcomes, however no practical biomarkers exist. This study examined whether eosinophils could be effective biomarkers for ≥grade 2 irAEs in RCC. METHODS We retrospectively analyzed 75 patients with RCC treated with ipilimumab and nivolumab between August 2018 and March 2021 in a multicenter study. Eosinophils were examined before and 2 weeks after treatment, and immediately after irAEs development. The optimal cut-off value for ≥grade 2 irAEs was determined by a receiver operating characteristic (ROC) curve. Univariate and multivariate analyses were undertaken to identify predictors of ≥grade 2 irAEs. RESULTS Two weeks after treatment, eosinophils were significantly upregulated in patients who had experienced ≥grade 2 irAEs than in those who had not experienced irAEs (mean, 5.7% vs. 3.2%; p < 0.05). The optimal cut-off value for eosinophils against ≥grade 2 irAEs was 3.0% (area under the curve = 0.69). In multivariate analyses, an eosinophil level ≥ 3.0% was a risk factor for ≥grade 2 irAEs (odds ratio 4.18, 95% confidence interval 1.16-15.1). The eosinophil level 2 weeks after treatment was upregulated by the onset of any type of irAEs including endocrine, gastrointestinal, pulmonary and skin disorders. CONCLUSIONS An increased eosinophil level 2 weeks after treatment might be an effective biomarker for ≥grade 2 irAEs in patients with RCC treated with ipilimumab and nivolumab.
Collapse
Affiliation(s)
- Yoshihiko Tasaki
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Nagoya, Japan
| | - Yosuke Sugiyama
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Nami Tomiyama
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Nagoya, Japan
| | - Taku Naiki
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Nagoya, Japan
| | - Toshiki Etani
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Nagoya, Japan
| | - Kazumi Taguchi
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Nagoya, Japan
| | - Nayuka Matsuyama
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Nagoya, Japan
| | - Yasuhito Sue
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Nagoya, Japan
| | - Yoshihisa Mimura
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Hiroki Kubota
- Department of Urology, Kainan Hospital, Yatomi, Aichi, Japan
| | - Yusuke Noda
- Department of Urology, Toyota Kosei Hospital, Toyota, Aichi, Japan
| | - Maria Aoki
- Department of Urology, Nagoya East Medical Center, Nagoya, Aichi, Japan
| | | | - Satoshi Nozaki
- Department of Urology, Nagoya Tokushukai General Hospital, Kasugai, Aichi, Japan
| | - Satoshi Kurokawa
- Department of Urology, Nagoya Tokushukai General Hospital, Kasugai, Aichi, Japan
| | - Atsushi Okada
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Nagoya, Japan
| | - Noriyasu Kawai
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Nagoya, Japan
| | - Takahiro Yasui
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Nagoya, Japan
| | - Kazunori Kimura
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| |
Collapse
|
37
|
Khawar MB, Gao G, Rafiq M, Shehzadi A, Afzal A, Abbasi MH, Sheikh N, Afzal N, Ashraf MA, Hamid SE, Shahzaman S, Kawish N, Sun H. Breaking down barriers: The potential of smarter CAR-engineered NK cells against solid tumors. J Cell Biochem 2023; 124:1082-1104. [PMID: 37566723 DOI: 10.1002/jcb.30460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/04/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
Natural killer (NK) cells are considered to be the foremost fighters of our innate immune system against foreign invaders and thus tend to promptly latch onto the virus-infected and tumor/cancerous cells, killing them through phagocytosis. At present, the application of genetically engineered Chimeric antigen receptor (CAR) receptors ensures a guaranteed optimistic response with NK cells and would not allow the affected cells to dodge or escape unchecked. Hence the specificity and uniqueness of CAR-NK cells over CAR-T therapy make them a better immunotherapeutic choice to reduce the load of trafficking of numerous tumor cells near the healthy cell populations in a more intact way than offered by CAR-T immunotherapy. Our review mainly focuses on the preclinical, clinical, and recent advances in clinical research trials and further strategies to achieve an augmented and efficient cure against solid tumors.
Collapse
Affiliation(s)
- Muhammad B Khawar
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental and, Translational Noncoding RNA Research, Yangzhou, China
- Department of Zoology, Applied Molecular Biology and Biomedicine Lab, University of Narowal, Narowal, Pakistan
| | - Guangzhong Gao
- Department of Physiatry, Haian Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nantong, Jiangsu, China
| | - Mussarat Rafiq
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Anila Shehzadi
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Ali Afzal
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | | | - Nadeem Sheikh
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Nimra Afzal
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | | | - Syeda E Hamid
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Sara Shahzaman
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Naseer Kawish
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Haibo Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental and, Translational Noncoding RNA Research, Yangzhou, China
| |
Collapse
|
38
|
Difoum F, Schernberg A, Vanquaethem H, Picchi H, Roy AL, Vuagnat P, Helissey C. Prognostic factors of toxicity of immune checkpoint inhibitors in nonsmall cell lung cancer and small cell lung cancer patients: The ToxImmune study. Cancer Rep (Hoboken) 2023; 6:e1760. [PMID: 36494190 PMCID: PMC10363797 DOI: 10.1002/cnr2.1760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/07/2022] [Accepted: 11/27/2022] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Immunotherapy alone or in combination has clearly improved the survival of patients with lung cancer. However, it may also be responsible for adverse events impacting these patients' quality of life. The ToxImmune study aims to identify prognostic factors that can help to predict immune-related adverse events. METHODS We included all patients aged 18 years and older who had received at least one dose of immune checkpoint inhibitors, with or without other therapy, between June 2015 and December 2020 and were diagnosed with nonsmall cell lung cancer or small-cell lung cancer. Patients' baseline demographic characteristics, biological blood markers, and imaging by PET-scanner were collected from electronic medical records. All adverse events (AEs) and immune-related AEs (irAEs) were recorded (Common Terminology Criteria For Adverse Events V.5.0). RESULTS Sixty-four patients were included, of whom 60 (94%) presented at least one irAE. The incidence of Common Terminology Criteria for Adverse Events (CTCAE) grade 2 and grade 3-4 was 34% and 8% respectively. Female sex, Primitive Tumor Standardized Uptake Value Max (SUVmax) <5, number of metastases ≥3 and immunotherapy received after the first line were found to be significant risk factors for immune-related adverse events. Based on the number of risk factors, the ToxImmune score predicts the risk of having a grade ≥2 adverse event (primitive tumor SUV ≥ 5 = 0 vs. primitive tumor SUV <5 = 1, number of metastases <3 = 0 vs. number of metastases ≥3 = 1 and L1 = 0 vs. L1 ≥ 1). The incidence of grade ≥2 adverse events was 20%, 55% and 90% with ToxImmune scores 0, 1 and = 2 respectively (p = .003). Median progression-free survival (PFS) times were 19.2 months, 6.64 months and 2.63 months for ToxImmune scores 0, 1 and = 2 respectively, p = .13. Median overall survival times were 22.6 months, 16.4 months and 9.8 months for ToxImmune scores 0, 1 and ≥2 respectively, p = .24. The disease control rate (DRR) was 78% in ToxIummune score 0 group, and 50% in ToxImmune score 1 and ≥2 groups (p = .363). CONCLUSION The ToxImmune score, which is grounded on objective clinical parameters, indicates that cases with a high score had an advanced threat of severe adverse events. The ToxImmune score could therefore be used in clinical practice to identify patients treated for lung cancer with immunotherapy and at risk of severe AE.
Collapse
Affiliation(s)
- Francoise Difoum
- Clinical Research unit, Military Hospital Begin, Saint-Mandé, France
| | | | - Hélène Vanquaethem
- Department of Internal Medicine, Military Hospital Begin, Saint-Mandé, France
| | - Hugo Picchi
- Department of Medical oncology, Military Hospital Begin, Saint-Mandé, France
| | - Audrey Le Roy
- Department of Medical oncology, Military Hospital Begin, Saint-Mandé, France
| | - Perrine Vuagnat
- Clinical Research unit, Military Hospital Begin, Saint-Mandé, France
| | - Carole Helissey
- Clinical Research unit, Military Hospital Begin, Saint-Mandé, France
- Department of Medical oncology, Military Hospital Begin, Saint-Mandé, France
| |
Collapse
|
39
|
Rodrigues M, Vanoni G, Loap P, Dubot C, Timperi E, Minsat M, Bazire L, Durdux C, Fourchotte V, Laas E, Pouget N, Castel-Ajgal Z, Marret G, Lesage L, Meseure D, Vincent-Salomon A, Lecompte L, Servant N, Vacher S, Bieche I, Malhaire C, Huchet V, Champion L, Kamal M, Amigorena S, Lantz O, Chevrier M, Romano E. Nivolumab plus chemoradiotherapy in locally-advanced cervical cancer: the NICOL phase 1 trial. Nat Commun 2023; 14:3698. [PMID: 37349318 PMCID: PMC10287640 DOI: 10.1038/s41467-023-39383-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 06/12/2023] [Indexed: 06/24/2023] Open
Abstract
Concurrent chemoradiotherapy (CRT) with blockade of the PD-1 pathway may enhance immune-mediated tumor control through increased phagocytosis, cell death, and antigen presentation. The NiCOL phase 1 trial (NCT03298893) is designed to determine the safety/tolerance profile and the recommended phase-II dose of nivolumab with and following concurrent CRT in 16 women with locally advanced cervical cancer. Secondary endpoints include objective response rate (ORR), progression free survival (PFS), disease free survival, and immune correlates of response. Three patients experience grade 3 dose-limiting toxicities. The pre-specified endpoints are met, and overall response rate is 93.8% [95%CI: 69.8-99.8%] with a 2-year PFS of 75% [95% CI: 56.5-99.5%]. Compared to patients with progressive disease (PD), progression-free (PF) subjects show a brisker stromal immune infiltrate, higher proximity of tumor-infiltrating CD3+ T cells to PD-L1+ tumor cells and of FOXP3+ T cells to proliferating CD11c+ myeloid cells. PF show higher baseline levels of PD-1 and ICOS-L on tumor-infiltrating EMRA CD4+ T cells and tumor-associated macrophages, respectively; PD instead, display enhanced PD-L1 expression on TAMs, higher peripheral frequencies of proliferating Tregs at baseline and higher PD-1 levels at week 6 post-treatment initiation on CD4 and CD8 T cell subsets. Concomitant nivolumab plus definitive CRT is safe and associated with encouraging PFS rates. Further validation in the subset of locally advanced cervical cancer displaying pre-existing, adaptive immune activation is warranted.
Collapse
Affiliation(s)
- Manuel Rodrigues
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France
| | - Giulia Vanoni
- Center for Cancer Immunotherapy, INSERM U932, PSL Research University, Institut Curie, Paris, France
| | - Pierre Loap
- Department of Radiation Oncology, Institut Curie, Paris & Saint Cloud, France
| | - Coraline Dubot
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France
| | - Eleonora Timperi
- Center for Cancer Immunotherapy, INSERM U932, PSL Research University, Institut Curie, Paris, France
| | - Mathieu Minsat
- Department of Radiation Oncology, Institut Curie, Paris & Saint Cloud, France
| | - Louis Bazire
- Department of Radiation Oncology, Institut Curie, Paris & Saint Cloud, France
| | - Catherine Durdux
- Hôpital Européen Georges Pompidou, Department of Radiation Oncology, Paris, France
| | | | - Enora Laas
- Service of Breast and Gynecologic Surgery, Institut Curie, Paris, France
| | - Nicolas Pouget
- Service of Breast and Gynecologic Surgery, Institut Curie, Paris, France
| | - Zahra Castel-Ajgal
- Department of Drug Development and Innovation, Institut Curie, Paris, France
| | - Gregoire Marret
- Department of Drug Development and Innovation, Institut Curie, Paris, France
| | - Laetitia Lesage
- Department of Pathology Institut Curie, Paris, France
- Centre d'Investigation Clinique Biothérapie, Institut Curie, Paris, France
| | - Didier Meseure
- Department of Pathology Institut Curie, Paris, France
- Centre d'Investigation Clinique Biothérapie, Institut Curie, Paris, France
| | - Anne Vincent-Salomon
- Department of Pathology Institut Curie, Paris, France
- Centre d'Investigation Clinique Biothérapie, Institut Curie, Paris, France
| | - Lolita Lecompte
- Institut Curie Bioinformatics Platform, INSERM U900, Mines ParisTech, Paris, 75005, France
| | - Nicolas Servant
- Institut Curie Bioinformatics Platform, INSERM U900, Mines ParisTech, Paris, 75005, France
| | - Sophie Vacher
- Pharmacogenomics Unit, Service of Genetics, Institut Curie, Paris, France
| | - Ivan Bieche
- Pharmacogenomics Unit, Service of Genetics, Institut Curie, Paris, France
| | | | - Virginie Huchet
- Department of Nuclear Medicine, Institut Curie, Paris, 75005, France
| | - Laurence Champion
- Department of Nuclear Medicine, Institut Curie, Paris, 75005, France
| | - Maud Kamal
- Department of Drug Development and Innovation, Institut Curie, Paris, France
| | - Sebastian Amigorena
- Center for Cancer Immunotherapy, INSERM U932, PSL Research University, Institut Curie, Paris, France
| | - Olivier Lantz
- Center for Cancer Immunotherapy, INSERM U932, PSL Research University, Institut Curie, Paris, France
| | - Marion Chevrier
- Service of Biostatistics, Institut Curie, Paris, 75005, France
| | - Emanuela Romano
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France.
- Center for Cancer Immunotherapy, INSERM U932, PSL Research University, Institut Curie, Paris, France.
| |
Collapse
|
40
|
Cao T, Zhou X, Wu X, Zou Y. Cutaneous immune-related adverse events to immune checkpoint inhibitors: from underlying immunological mechanisms to multi-omics prediction. Front Immunol 2023; 14:1207544. [PMID: 37497220 PMCID: PMC10368482 DOI: 10.3389/fimmu.2023.1207544] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/05/2023] [Indexed: 07/28/2023] Open
Abstract
The development of immune checkpoint inhibitors (ICIs) has dramatically altered the landscape of therapy for multiple malignancies, including urothelial carcinoma, non-small cell lung cancer, melanoma and gastric cancer. As part of their anti-tumor properties, ICIs can enhance susceptibility to inflammatory side effects known as immune-related adverse events (irAEs), in which the skin is one of the most commonly and rapidly affected organs. Although numerous questions still remain unanswered, multi-omics technologies have shed light into immunological mechanisms, as well as the correlation between ICI-induced activation of immune systems and the incidence of cirAE (cutaneous irAEs). Therefore, we reviewed integrated biological layers of omics studies combined with clinical data for the prediction biomarkers of cirAEs based on skin pathogenesis. Here, we provide an overview of a spectrum of dermatological irAEs, discuss the pathogenesis of this "off-tumor toxicity" during ICI treatment, and summarize recently investigated biomarkers that may have predictive value for cirAEs via multi-omics approach. Finally, we demonstrate the prognostic significance of cirAEs for immune checkpoint blockades.
Collapse
|
41
|
Van Mol P, Donders E, Lambrechts D, Wauters E. Immune checkpoint biology in health & disease: Immune checkpoint biology and autoimmunity in cancer patients. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 382:181-206. [PMID: 38225103 DOI: 10.1016/bs.ircmb.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Immune checkpoints (ICs) play a central role in maintaining immune homoeostasis. The discovery that tumours use this physiological mechanism to avoid elimination by the immune system, opened up avenues for therapeutic targeting of ICs as a novel way of treating cancer. However, this therapy a new array of autoimmune side effects, termed immune-related adverse events (irAEs). In this narrative review, we first recapitulate the physiological function of ICs that are approved targets for cancer immunotherapy (CTLA-4, PD-(L)1 and LAG-3), as the groundwork to critically discuss current knowledge on irAEs. Specifically, we summarize clinical aspects and examine a molecular classification and predisposing factors of irAEs. Finally, we discuss irAE treatment, particularly emphasizing how molecular knowledge is changing the current treatment paradigm.
Collapse
Affiliation(s)
- Pierre Van Mol
- VIB - CCB Laboratory of Translational Genetics, KU Leuven, Leuven, Belgium; Pneumology - Respiratory Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Elena Donders
- VIB - CCB Laboratory of Translational Genetics, KU Leuven, Leuven, Belgium; Pneumology - Respiratory Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Diether Lambrechts
- VIB - CCB Laboratory of Translational Genetics, KU Leuven, Leuven, Belgium
| | - Els Wauters
- Pneumology - Respiratory Oncology, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
42
|
Sun L, Meng C, Zhang X, Gao J, Wei P, Zhang J, Zhang Z. Management and prediction of immune-related adverse events for PD1/PDL-1 immunotherapy in colorectal cancer. Front Pharmacol 2023; 14:1167670. [PMID: 37188271 PMCID: PMC10176603 DOI: 10.3389/fphar.2023.1167670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Programmed cell death protein (PD-1) is an important immunosuppressive molecule, which can inhibit interaction between PD-1 and its ligand PD-L1, further enhancing the T cell response and anti-tumor activity, which is called immune checkpoint blockade. Immunotherapy, represented by immune checkpoint inhibitors, has opened up a new era of tumor treatment and is gradually being applied to colorectal cancer recently. Immunotherapy was reported could achieve a high objective response rate (ORR) for colorectal cancer with high microsatellite instability (MSI), thus opening up a new era of colorectal cancer immunotherapy. Along with the increasing use of PD1 drugs in colorectal cancer, we should pay more attention to the adverse effects of these immune drugs while seeing the hope. Immune-related adverse events (irAEs) caused by immune activation and immune homeostasis during anti-PD-1/PD-L1 therapy can affect multi-organ and even be fatal in serious cases. Therefore, understanding irAEs is essential for their early detection and appropriate management. In this article, we review the irAEs that occur during the treatment of colorectal cancer patients with PD-1/PD-L1 drugs, analyze the current controversies and challenges, and point out future directions that should be explored, including exploring efficacy predictive markers and optimizing the paradigm of individualized immunotherapy.
Collapse
Affiliation(s)
- Liting Sun
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Cong Meng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xiao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jiale Gao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Pengyu Wei
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jie Zhang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
43
|
Athale J, Broderick K, Wu X, Grossman S. Evidence for Persistent Radiation-Related Injury to the Immune System in Patients With Local Breast Cancer. Int J Radiat Oncol Biol Phys 2023; 115:1192-1196. [PMID: 36528228 PMCID: PMC10084788 DOI: 10.1016/j.ijrobp.2022.11.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 11/04/2022] [Accepted: 11/11/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Janhavi Athale
- Critical Care Medicine, Mayo Clinic Arizona, Phoenix, Arizona.
| | | | - Xiaojun Wu
- Johns Hopkins Hospital, Baltimore, Maryland
| | - Stuart Grossman
- Brain Cancer Research Program, Sidney Kimmel Cancer Center, Baltimore, Maryland
| |
Collapse
|
44
|
Yang Y, Liu H, Chen Y, Xiao N, Zheng Z, Liu H, Wan J. Liquid biopsy on the horizon in immunotherapy of non-small cell lung cancer: current status, challenges, and perspectives. Cell Death Dis 2023; 14:230. [PMID: 37002211 PMCID: PMC10066332 DOI: 10.1038/s41419-023-05757-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 04/03/2023]
Abstract
Non-small cell lung cancer (NSCLC) is one of the most threatening malignancies to human health and life. In most cases, patients with NSCLC are already at an advanced stage when they are diagnosed. In recent years, lung cancer has made great progress in precision therapy, but the efficacy of immunotherapy is unstable, and its response rate varies from patient to patient. Several biomarkers have been proposed to predict the outcomes of immunotherapy, such as programmed cell death-ligand 1 (PD-L1) expression and tumor mutational burden (TMB). Nevertheless, the detection assays are invasive and demanding on tumor tissue. To effectively predict the outcomes of immunotherapy, novel biomarkers are needed to improve the performance of conventional biomarkers. Liquid biopsy is to capture and detect circulating tumor cells (CTCs), circulating tumor DNA (ctDNA) and exosomes in body fluids, such as blood, saliva, urine, pleural fluid and cerebrospinal fluid as samples from patients, so as to make analysis and diagnosis of cancer and other diseases. The application of liquid biopsy provides a new possible solution, as it has several advantages such as non-invasive, real-time dynamic monitoring, and overcoming tumor heterogeneity. Liquid biopsy has shown predictive value in immunotherapy, significantly improving the precision treatment of lung cancer patients. Herein, we review the application of liquid biopsy in predicting the outcomes of immunotherapy in NSCLC patients, and discuss the challenges and future directions in this field.
Collapse
Affiliation(s)
- Ying Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Youming Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Nan Xiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhaoyang Zheng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Hongchun Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
45
|
Les I, Martínez M, Pérez-Francisco I, Cabero M, Teijeira L, Arrazubi V, Torrego N, Campillo-Calatayud A, Elejalde I, Kochan G, Escors D. Predictive Biomarkers for Checkpoint Inhibitor Immune-Related Adverse Events. Cancers (Basel) 2023; 15:1629. [PMID: 36900420 PMCID: PMC10000735 DOI: 10.3390/cancers15051629] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Immune-checkpoint inhibitors (ICIs) are antagonists of inhibitory receptors in the immune system, such as the cytotoxic T-lymphocyte-associated antigen-4, the programmed cell death protein-1 and its ligand PD-L1, and they are increasingly used in cancer treatment. By blocking certain suppressive pathways, ICIs promote T-cell activation and antitumor activity but may induce so-called immune-related adverse events (irAEs), which mimic traditional autoimmune disorders. With the approval of more ICIs, irAE prediction has become a key factor in improving patient survival and quality of life. Several biomarkers have been described as potential irAE predictors, some of them are already available for clinical use and others are under development; examples include circulating blood cell counts and ratios, T-cell expansion and diversification, cytokines, autoantibodies and autoantigens, serum and other biological fluid proteins, human leucocyte antigen genotypes, genetic variations and gene profiles, microRNAs, and the gastrointestinal microbiome. Nevertheless, it is difficult to generalize the application of irAE biomarkers based on the current evidence because most studies have been retrospective, time-limited and restricted to a specific type of cancer, irAE or ICI. Long-term prospective cohorts and real-life studies are needed to assess the predictive capacity of different potential irAE biomarkers, regardless of the ICI type, organ involved or cancer site.
Collapse
Affiliation(s)
- Iñigo Les
- Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Autoimmune Diseases Unit, Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Inflammatory and Immune-Mediated Diseases Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - Mireia Martínez
- Osakidetza Basque Health Service, Department of Medical Oncology, Araba University Hospital, 01009 Vitoria-Gasteiz, Spain
- Lung Cancer Research Group, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - Inés Pérez-Francisco
- Breast Cancer Research Group, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - María Cabero
- Clinical Trials Platform, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - Lucía Teijeira
- Medical Oncology Department, Navarre University Hospital, 31008 Pamplona, Spain
| | - Virginia Arrazubi
- Medical Oncology Department, Navarre University Hospital, 31008 Pamplona, Spain
| | - Nuria Torrego
- Osakidetza Basque Health Service, Department of Medical Oncology, Araba University Hospital, 01009 Vitoria-Gasteiz, Spain
- Lung Cancer Research Group, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - Ana Campillo-Calatayud
- Inflammatory and Immune-Mediated Diseases Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - Iñaki Elejalde
- Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Autoimmune Diseases Unit, Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Inflammatory and Immune-Mediated Diseases Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - Grazyna Kochan
- Oncoimmunology Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - David Escors
- Oncoimmunology Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| |
Collapse
|
46
|
van der Kooij MK, Joosse A, Suijkerbuijk KPM, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Boers-Sonderen MJ, van den Eertwegh AJM, de Groot JWB, Haanen JBAG, Hospers GAP, Piersma D, van Rijn RS, van der Veldt AAM, Vreugdenhil G, Westgeest HM, Wouters MWJM, Dekkers OM, Kapiteijn E. Failure to validate existing clinical prediction scale for response to PD-1 monotherapy in advanced melanoma in national cohort study. Br J Cancer 2023; 128:707-710. [PMID: 36564561 PMCID: PMC9977837 DOI: 10.1038/s41416-022-02088-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 11/04/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Monique K van der Kooij
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, PO box 9600, 2300 RC, Leiden, The Netherlands
| | - Arjen Joosse
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Maureen J B Aarts
- Department of Medical Oncology, Maastricht University Medical Center, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands
| | | | - Christian U Blank
- Department of Medical Oncology, Netherlands Cancer Institute- Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Marye J Boers-Sonderen
- Department of Medical Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6500 HB, Nijmegen, The Netherlands
| | - Alfonsus J M van den Eertwegh
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, 1081 HZ, Amsterdam, The Netherlands
| | | | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute- Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Geke A P Hospers
- Department of Medical Oncology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Djura Piersma
- Department of Medical Oncology, Medisch Spectrum Twente, Koningsplein 1, 7512 KZ, Enschede, The Netherlands
| | - Rozemarijn S van Rijn
- Department of Medical Oncology, Medical Center Leeuwarden, Henri Dunantweg 2, 8934 AD, Leeuwarden, The Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands
| | - Gerard Vreugdenhil
- Department of Medical Oncology, Maxima Medical Center, de Run 4600, 5500 MB, Veldhoven, The Netherlands
| | - Hans M Westgeest
- Department of Medical Oncology, Amphia Ziekenhuis, Langendijk 175, 4819 EV, Breda, The Netherlands
| | - Michel W J M Wouters
- Dutch Institute for Clinical Auditing, Rijnsburgerweg 10, 2333 AA, Leiden, The Netherlands
- Department of Surgical Oncology, Netherlands Cancer Institute- Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Olaf M Dekkers
- Department of Clinical Epidemiology, Leiden University Medical Center, Albinusdreef 2, PO box 9600, 2300 RC, Leiden, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, PO box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
47
|
Kang MK. Implications of recent neoadjuvant clinical trials on the future practice of radiotherapy in locally advanced rectal cancer. World J Gastroenterol 2023; 29:1011-1025. [PMID: 36844136 PMCID: PMC9950859 DOI: 10.3748/wjg.v29.i6.1011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/08/2022] [Accepted: 01/29/2023] [Indexed: 02/10/2023] Open
Abstract
Over the last two decades, the standard treatment for locally advanced rectal cancer (LARC) has been neoadjuvant chemoradiotherapy plus total mesorectal excision followed by adjuvant chemotherapy. Total neoadjuvant treatment (TNT) and immunotherapy are two major issues in the treatment of LARC. In the two latest phase III randomized controlled trials (RAPIDO and PRODIGE23), the TNT approach achieved higher rates of pathologic complete response and distant metastasis-free survival than conventional chemoradiotherapy. Phase I/II clinical trials have reported promising response rates to neoadjuvant (chemo)-radiotherapy combined with immunotherapy. Accordingly, the treatment paradigm for LARC is shifting toward methods that increase the oncologic outcomes and organ preservation rate. However, despite the progress of these combined modality treatment strategies for LARC, the radiotherapy details in clinical trials have not changed significantly. To guide future radiotherapy for LARC with clinical and radiobiological evidence, this study reviewed recent neoadjuvant clinical trials evaluating TNT and immunotherapy from a radiation oncologist's perspective.
Collapse
Affiliation(s)
- Min Kyu Kang
- Department of Radiation Oncology, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
- Department of Radiation Oncology, Kyungpook National University Chilgok Hospital, Daegu 40414, South Korea
| |
Collapse
|
48
|
Effect of lymphoid volume irradiation on radiation-induced lymphopenia in head and neck cancers. Cancer Radiother 2023; 27:145-153. [PMID: 36759240 DOI: 10.1016/j.canrad.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/21/2022] [Accepted: 08/25/2022] [Indexed: 02/10/2023]
Abstract
PURPOSE Radiotherapy induces significant and prolonged lymphopenia in head and neck cancer patients with poorer outcomes and reduced survival. Irradiated volumes may be correlated with lymphopenia with a potential impact on immunotherapy efficacy. We assessed associations between volumes treated with radiotherapy and the nadir of the lymphocyte count in patients with head and neck cancer. MATERIALS AND METHODS We conducted a monocentric retrospective study in patients with head and neck cancer treated with radiation. Univariate analysis used regression analysis to model nadir lymphocyte count and radiotherapy volumes; multivariate analysis then modelled factors associated with nadir lymphocyte count. RESULTS Of the 77 included patients, 97% presented lymphopenia during radiotherapy with an average nadir of 431 cells/mm3 at a median of 40 days after the beginning of treatment. The volume of high-risk radiotherapy and gross tumour volume were correlated with nadir lymphocyte count with a Spearman coefficient of -0.267 (P=0.019) and -0.387 (P=0.001), respectively. After multivariate linear regression, high-risk radiotherapy was significantly associated with nadir lymphocyte count with a regression coefficient of -0.32 (per cubic centimetre) [95% CI=-0.60; -0.03] (P=0.028). CONCLUSION High-risk radiotherapy was significantly associated with nadir lymphocyte count in patients with head and neck cancer treated with radiation. Sparing lymphoid volumes from irradiation by elective nodal irradiation or proton therapy may limit lymphopenia and needs to be investigated in combination with immunotherapy.
Collapse
|
49
|
Yamamoto T, Morooka H, Ito T, Ishigami M, Mizuno K, Yokoyama S, Yamamoto K, Imai N, Ishizu Y, Honda T, Yokota K, Hase T, Maeda O, Hashimoto N, Ando Y, Akiyama M, Kawashima H. Clustering using unsupervised machine learning to stratify the risk of immune-related liver injury. J Gastroenterol Hepatol 2023; 38:251-258. [PMID: 36302734 DOI: 10.1111/jgh.16038] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/26/2022] [Accepted: 10/22/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM Immune-related liver injury (liver-irAE) is a clinical problem with a potentially poor prognosis. METHODS We retrospectively collected clinical data from patients treated with immune checkpoint inhibitors between September 2014 and December 2021 at the Nagoya University Hospital. Using an unsupervised machine learning method, the Gaussian mixture model, to divide the cohort into clusters based on inflammatory markers, we investigated the cumulative incidence of liver-irAEs in these clusters. RESULTS This study included a total of 702 patients. Among them, 492 (70.1%) patients were male, and the mean age was 66.6 years. During the mean follow-up period of 423 days, severe liver-irAEs (Common Terminology Criteria for Adverse Events grade ≥ 3) occurred in 43 patients. Patients were divided into five clusters (a, b, c, d, and e). The cumulative incidence of liver-irAE was higher in cluster c than in cluster a (hazard ratio [HR]: 13.59, 95% confidence interval [CI]: 1.70-108.76, P = 0.014), and overall survival was worse in clusters c and d than in cluster a (HR: 2.83, 95% CI: 1.77-4.50, P < 0.001; HR: 2.87, 95% CI: 1.47-5.60, P = 0.002, respectively). Clusters c and d were characterized by high temperature, C-reactive protein, platelets, and low albumin. However, there were differences in the prevalence of neutrophil count, neutrophil-to-lymphocyte ratio, and liver metastases between both clusters. CONCLUSIONS The combined assessment of multiple markers and body temperature may help stratify high-risk groups for developing liver-irAE.
Collapse
Affiliation(s)
- Takafumi Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hikaru Morooka
- Department of Emergency and Critical Care Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masatoshi Ishigami
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuyuki Mizuno
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinya Yokoyama
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenta Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norihiro Imai
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoji Ishizu
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Honda
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji Yokota
- Department of Dermatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsunari Hase
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Osamu Maeda
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Naozumi Hashimoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Masashi Akiyama
- Department of Dermatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
50
|
Shalit A, Sarantis P, Koustas E, Trifylli EM, Matthaios D, Karamouzis MV. Predictive Biomarkers for Immune-Related Endocrinopathies following Immune Checkpoint Inhibitors Treatment. Cancers (Basel) 2023; 15:375. [PMID: 36672324 PMCID: PMC9856539 DOI: 10.3390/cancers15020375] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
In recent years, in the context of the increase in the life expectancy of cancer patients, special attention has been given to immunotherapy and, indeed, to immune checkpoint inhibitors. The use of immune checkpoint inhibitors has increased rapidly, and approximately 40% of cancer patients are eligible for this treatment. Although their impact is valuable on cancer treatment, immune checkpoint inhibitors come with side effects, known as immune-related adverse effects. These can affect many systems, including cutaneous, musculoskeletal, cardiovascular, gastrointestinal, endocrine, neural, and pulmonary systems. In this review, we focus on immune-related endocrinopathies that affect around 10% of all treated patients. Endocrine dysfunctions can manifest as hypophysitis, thyroid dysfunction, hypoparathyroidism, insulin-deficient diabetes mellitus, and primary adrenal insufficiency. Currently, there are multiple ongoing clinical trials that aim to identify possible predictive biomarkers for immune-related adverse effects. The design of those clinical trials relies on collecting a variety of biological specimens (tissue biopsy, blood, plasma, saliva, and stool) at baseline and regular intervals during treatment. In this review, we present the predictive biomarkers (such as antibodies, hormones, cytokines, human leukocyte antigens, and eosinophils) that could potentially be utilized in clinical practice in order to predict adverse effects and manage them appropriately.
Collapse
Affiliation(s)
- Almog Shalit
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Panagiotis Sarantis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Evangelos Koustas
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- First Department of Internal Medicine, 417 Army Share Fund Hospital, 11521 Athens, Greece
| | - Eleni-Myrto Trifylli
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- First Department of Internal Medicine, 417 Army Share Fund Hospital, 11521 Athens, Greece
| | | | - Michalis V. Karamouzis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|