1
|
Zahnreich S, Bhatti A, Ahmad B, Drabke S, Kaufmann J, Schmidberger H. Effects of Cisplatin on the Radiation Response and DNA Damage Markers in Peripheral Blood Lymphocytes Ex Vivo. Cells 2025; 14:682. [PMID: 40422185 DOI: 10.3390/cells14100682] [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: 04/01/2025] [Revised: 04/30/2025] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Platinum-based radiochemotherapy is associated with hematologic side effects, impacting patient outcomes. However, the clinical mechanisms of cisplatin and its interaction with ionizing radiation (IR), including in biodosimetry for radiotherapy, have not yet been fully clarified. For this purpose, healthy donors' peripheral blood lymphocytes (PBLs) were pretreated with cisplatin in a pulse (1-4 h) or continuous (24 h) regimen followed by X-rays. DNA damage was assessed as DNA double-strand breaks using repair foci of γH2AX and 53BP1 after 0.5 h and 24 h in G1 PBLs and a proliferation-based cytokinesis-block micronucleus assay. Additionally, cell death and proliferation activity were measured. Unlike a 1 h pulse, a 24 h cisplatin pretreatment caused a concentration-dependent increase in cisplatin-induced foci while decreasing IR-induced foci, especially 24 h after irradiation. This was accompanied by increased apoptosis, with cisplatin and IR having additive effects. Both genotoxins alone caused a dose-dependent increase in micronuclei, while cisplatin significantly reduced binuclear cells, especially after the 24 h treatment, leading to lower micronuclei frequencies post-irradiation. Our results show that prolonged cisplatin exposure, even at low concentrations, impacts the vitality and division activity of PBLs, with significantly stronger effects post-irradiation. This has major implications and must be considered for the detection of DNA damage-associated biomarkers in PBLs used in clinical prediction or biodosimetry during radiotherapy.
Collapse
Affiliation(s)
- Sebastian Zahnreich
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Aisha Bhatti
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Barea Ahmad
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Sophia Drabke
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Justus Kaufmann
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Heinz Schmidberger
- Department of Radiation Oncology and Radiation Therapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| |
Collapse
|
2
|
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
|
3
|
Fang C, He X, Tang F, Wang Z, Pan C, Zhang Q, Wu J, Wang Q, Liu D, Zhang Y. Where lung cancer and tuberculosis intersect: recent advances. Front Immunol 2025; 16:1561719. [PMID: 40242762 PMCID: PMC11999974 DOI: 10.3389/fimmu.2025.1561719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Lung cancer (LC) and tuberculosis (TB) represent two major global public health issues. Prior evidence has suggested a link between TB infection and an increased risk of LC. As advancements in LC treatment have led to extended survival rates for LC patients, the co-occurrence of TB and LC has grown more prevalent and poses novel clinical challenges. The intricate molecular mechanisms connecting TB and LC are closely intertwined and many issues remain to be addressed. This review focuses on resemblance between the immunosuppression in tumor and granuloma microenvironments, exploring immunometabolism, cell plasticity, inflammatory signaling pathways, microbiomics, and up-to-date information derived from spatial multi-omics between TB and LC. Furthermore, we outline immunization-related molecular mechanisms underlying these two diseases and propose future research directions. By discussing recent advances and potential targets, this review aims to establish a foundation for developing future therapeutic strategies targeting LC with concurrent TB infection.
Collapse
Affiliation(s)
- Chunju Fang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Xuanlu He
- School of Clinical Medicine, Zunyi Medical University, Zunyi, China
| | - Fei Tang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Zi Wang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Cong Pan
- School of Biological Sciences, Guizhou Education University, Guiyang, China
- Translational Medicine Research Center, eBond Pharmaceutical Technology Co., Ltd., Chengdu, China
| | - Qi Zhang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jing Wu
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Qinglan Wang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Daishun Liu
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Yu Zhang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
- National Health Commission Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People’s Hospital, Guiyang, China
| |
Collapse
|
4
|
Paczona VR, Végváry Z, Kelemen G, Dobi Á, Borzási E, Varga L, Cserháti A, Csomor A, Radics B, Dósa S, Balázsfi M, Fodor E, Borzák F, Puskás Á, Varga Z, Oláh J, Hideghéty K. Magnetic resonance imaging in glioblastoma radiotherapy - beyond treatment adaptation. Phys Imaging Radiat Oncol 2025; 34:100754. [PMID: 40231225 PMCID: PMC11994382 DOI: 10.1016/j.phro.2025.100754] [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: 11/02/2024] [Revised: 03/03/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Background and Purpose The treatment of glioblastoma remains a challenging task for modern radiation oncology. Adaptive radiotherapy potentially improves local control and reduces toxicity to healthy brain tissue. The purpose of the study was to examine the safety of adaptive radiotherapy in glioblastoma, using a margin-reduction approach based on an interim magnetic resonance image (MRI). Furthermore, it aimed to identify radiomorphological features that may correlate with disease outcome. Materials and Methods 108 glioblastoma patients receiving standard chemoradiotherapy underwent repeated MRI after 40 Gy. The images were compared to the pre-radiotherapy MRI, based on the following criteria: midline shift, perifocal edema, contrast enhancement, ventricular compression, new lesion outside the radiation field, gross tumor volume (GTV) and planning target volume (PTV) size. Target volumes were adjusted by taking into consideration the new intracranial conditions and the remaining 20 Gy was delivered. Statistical analysis consisted of the comparison of the radiomorphological features to overall and progression free survival. Results Increased or unchanged contrast enhancement (HR: 2.11 and 1.18 consecutively) and ventricular compression (HR: 13.58 and 2.53) on the interim MRI resulted in significantly poorer survival. GTV size (initial: 61.4 [3.8-170.9], adapted: 45.3 [0-206.8] cm3) reduction (absolute: -16.2 [-115.3-115.5] cm3, relative: -24.5 [-100-258.9] %) also had demonstrable impact on survival. Changes in PTV, however, did not significantly correlate with survival. Conclusions By reducing PTV based on an interim MRI, we achieved substantial sparing of critical normal tissues, without compromising survival. The established evaluation categories can facilitate the systematic review of interim MRI findings.
Collapse
Affiliation(s)
- Viktor R. Paczona
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Zoltán Végváry
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Gyöngyi Kelemen
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Ágnes Dobi
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Emőke Borzási
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Linda Varga
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Adrienne Cserháti
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Angéla Csomor
- Department of Radiology, University of Szeged. 6 Semmelweis utca, Szeged 6720, Hungary
| | - Bence Radics
- Department of Pathology, University of Szeged. 2 Állomás utca, Szeged 6725, Hungary
| | - Sándor Dósa
- Department of Pathology, University of Szeged. 2 Állomás utca, Szeged 6725, Hungary
| | - Márton Balázsfi
- Department of Neurosurgery, University of Szeged. 6 Semmelweis utca, Szeged 6720, Hungary
| | - Emese Fodor
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Ferenc Borzák
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Árpád Puskás
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Zoltán Varga
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Judit Oláh
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| | - Katalin Hideghéty
- Department of Oncotherapy, University of Szeged. 12 Korányi fasor, Szeged 6720, Hungary
| |
Collapse
|
5
|
Cui K, Zhang S, Du Y, Chai Y, Liang M, Hu S, Li J. Lower pre-conditioning absolute lymphocyte counts are associated with worse outcomes in haploidentical stem cell transplantation with myeloablative regimen in children. Front Immunol 2025; 16:1552263. [PMID: 40201172 PMCID: PMC11975963 DOI: 10.3389/fimmu.2025.1552263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Background Anti-thymocyte globulin (ATG) is frequently administered for preventing graft-versus-host disease (GVHD) in allogeneic hematopoietic stem cell transplantation (allo-HSCT). In patients with low absolute lymphocyte count (ALC) before conditioning, weight-based dosing of ATG may cause overexposure, negatively impacting prognosis. Method Clinical data of patients with hematological malignancies undergoing haploidentical HSCT (haplo-HSCT) at the Children's Hospital of Soochow University from January 2020 to June 2023 were collected. This study primarily aims to investigate the association between pre-conditioning ALC and clinical outcomes in pediatric acute leukemia or myelodysplastic syndromes patients receiving myeloablative haplo-HSCT. Results We included 130 patients treated at the Children's Hospital of Soochow University from January 2020 to June 2023. According to the cutoff of 500/μl, patients were divided into high and low ALC groups. Patients in the high ALC group experienced a higher incidence of II-IV acute GVHD (30.2% versus 13.6%, P = 0.034), 3-year overall survival (OS) and relapse-free survival (RFS) rates (OS: 88.5% ± 3.7% versus 66.9% ± 7.9%, P = 0.013; RFS: 81.4% ± 4.1% versus 56.5% ± 8.1%, P < 0.001), and lower cumulative incidence of relapse (11.3% versus 27.4%, P = 0.013). Pre-conditioning ALC < 500/μl independently predicted worse OS, RFS, and higher relapse risk in multivariate analysis. However, there was no significant difference in immune reconstitution between the two groups. Conclusion Pre-conditioning ALC was a significant prognostic factor in pediatric patients undergoing myeloablative haplo-HSCT. Further research is needed to explore whether pre-conditioning ALC can serve as a reference for adjusting ATG dosing.
Collapse
Affiliation(s)
- Kai Cui
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou, China
| | - Senlin Zhang
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou, China
| | - Yueke Du
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou, China
| | - Yutan Chai
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou, China
| | - Mingchu Liang
- Department of Nephrology and Immunology, Children’s Hospital of Soochow University, Suzhou, China
| | - Shaoyan Hu
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou, China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, China
| | - Jie Li
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou, China
| |
Collapse
|
6
|
Manzar GS, Wu SY, Dudzinski SO, Cha EE, Yoder AK, Corrigan KL, Nasr LF, Sallard G, Ahmed S, Fayad LE, Chihara D, Nair R, Westin JR, Daher M, Neelapu SS, Nastoupil LJ, Gunther JR, Pinnix CC, Dabaja BS, Strati P, Fang PQ. Characterization of Lymphopenia and Correlating the Risk of Cytopenias With Dose and Bone Marrow Volume Irradiated in Aggressive B Cell Lymphoma Patients Bridged With Radiation Therapy for Chimeric Antigen Receptor-T Cell Therapy. Int J Radiat Oncol Biol Phys 2025; 121:1011-1025. [PMID: 39303997 DOI: 10.1016/j.ijrobp.2024.09.023] [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: 03/22/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE The impact of bridging radiation therapy (bRT) for chimeric antigen receptor (CAR) T-cell therapy on absolute lymphocyte count (ALC) kinetics and treatment outcome is unknown. METHODS AND MATERIALS We retrospectively reviewed adults with relapsed/refractory aggressive large B cell lymphoma who received bRT before CD-19 CAR-T between November 2017 and April 2023. The change in ALC (ALC Δ RT) was computed by subtracting ALC pre- and post-bRT. Percent bone marrow (%BM) irradiated was calculated by estimating skeletal BM distribution. Progression-free survival (PFS), disease-specific survival (DSS), and overall survival (OS) were modeled via Kaplan-Meier. RESULTS Fifty-one patients received bRT, of which 13 (25.5%) had bulky disease (≥7.5 cm). The median bRT dose was 30 Gy (range, 4-48 Gy); 26 patients (51%) received ≥30 Gy. Thirty-one patients (61%) received bRT comprehensively to all disease sites. The median cumulative %BM irradiated was 5.05% (range, 0-50%). At a median follow-up of 10.3 months (95% CI, 7.7-16.4), the 1-year OS, PFS, and DSS rates were 80% (95% CI, 66-99), 78% (64-87), and 82% (68-90), respectively. The incidence of grade ≥3 lymphopenia was 33% pre-RT and 68% post-RT, but recovered to 43% at the conditioning chemotherapy timepoint. There was no correlation between post-RT grade ≥3 lymphopenia and the receipt of comprehensive bRT, combined modality bridging, ≥30 Gy bRT, or bRT to ≥15% of BM (all P > .2). Among patients with grade 0-2 lymphopenia pre-RT, increased conversion to grade ≥3 lymphopenia post-RT correlated with comprehensive or ≥30 Gy bRT, but these factors did not impair ALC recovery at conditioning chemotherapy. There was no association between ALC Δ RT or post-RT ALC with 30 or 90 day response (P > .25), DSS, PFS, or OS (P > .3). CONCLUSIONS Lymphocyte change during bRT is not associated with CAR-T outcomes. Persistent cytopenia risk after bRT is not associated with bRT to ≥30 Gy, ≥15% of BM, or comprehensive coverage. Although bRT can be delivered safely, we urge careful treatment planning when incorporating into pre-CAR-T regimens.
Collapse
MESH Headings
- Humans
- Lymphopenia/etiology
- Male
- Middle Aged
- Female
- Retrospective Studies
- Bone Marrow/radiation effects
- Bone Marrow/pathology
- Adult
- Aged
- Radiotherapy Dosage
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Lymphocyte Count
- Receptors, Chimeric Antigen/therapeutic use
- Lymphoma, Large B-Cell, Diffuse/radiotherapy
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Progression-Free Survival
- Aged, 80 and over
- Young Adult
- Cytopenia
Collapse
Affiliation(s)
- Gohar S Manzar
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Susan Y Wu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephanie O Dudzinski
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elaine E Cha
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alison K Yoder
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kelsey L Corrigan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lewis F Nasr
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Sairah Ahmed
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luis E Fayad
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dai Chihara
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ranjit Nair
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason R Westin
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - May Daher
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sattva S Neelapu
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Loretta J Nastoupil
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jillian R Gunther
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chelsea C Pinnix
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bouthaina S Dabaja
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paolo Strati
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Penny Q Fang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
7
|
Bott R, Zylstra J, Knight W, Whyte GP, Lane AM, Moss C, Browning M, Lagergren J, Van Hemelrijck M, Davies AR. Prehabilitation of Patients With Oesophageal Malignancy Undergoing Peri-Operative Treatment (Pre-EMPT): Outcomes From a Prospective Controlled Trial. J Surg Oncol 2025. [PMID: 39878357 DOI: 10.1002/jso.28079] [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: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 01/31/2025]
Abstract
BACKGROUND The Pre-EMPT study aimed to determine if structured exercise could reduce length of stay, post-operative complications and improve fitness and health-related quality of life (HQRL) in patients undergoing neoadjuvant chemotherapy (NAC) and oesophagectomy. METHODS A prospective non-randomised trial compared a standard care pathway (control) to a structured prehabilitation exercise programme (intervention) commenced before NAC and surgery for oesophageal adenocarcinoma. Length of hospital stay and post-operative complications were recorded. Cardiopulmonary exercise testing (CPEX), body composition analyses, lymphocyte levels and HRQL questionnaires were performed at multiple time points. RESULTS Median length of stay was similar in both groups. There were 6 versus 11 complications observed (intervention vs control p = 0.086). Cardio-pulmonary fitness (VO2peak) declined after NAC, but less in the intervention group (intervention -13.54% vs control -21.40%, p = 0.02). Body composition improved in the intervention group (FMi/FFMi -5.5% intervention, 10.7% control p = 0.043). Performance, cognitive, sleep and emotional function scores improved following NAC in the intervention group. Lymphocyte subsets increased in the intervention group compared to the control group after chemotherapy (p = 0.034). Chemotherapy response was improved in the intervention group (p = 0.022). CONCLUSION A structured exercise programme may mitigate cardiopulmonary deconditioning, reduce sarcopenia and offset lymphopenia, during chemotherapy, in patients undergoing NAC and oesophagectomy.
Collapse
Affiliation(s)
- R Bott
- Department of Upper GI and General Surgery, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - J Zylstra
- Department of Upper GI and General Surgery, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
- Sport and Exercise Sciences, Faculty of Science, Liverpool John Moore's University, Liverpool, United Kingdom
| | - W Knight
- Department of Upper GI and General Surgery, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - G P Whyte
- Sport and Exercise Sciences, Faculty of Science, Liverpool John Moore's University, Liverpool, United Kingdom
- Centre for Health and Human Performance, London, United Kingdom
| | - A M Lane
- University of Wolverhampton Research Centre for Sport, Exercise and Performance (RCSEP), Wolverhampton, United Kingdom
| | - C Moss
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - M Browning
- Department of Anaesthesia and Intensive Care, Maidstone and Tunbridge Wells NHS Trust, Maidstone, United Kingdom
| | - J Lagergren
- Department of Upper GI and General Surgery, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - M Van Hemelrijck
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - A R Davies
- Department of Upper GI and General Surgery, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
8
|
Luo D, Zhong Q, Yue H, Wang J, Liang Q, Liu W, Zhu X. The predictors of lymphopenia and its effects on survival in locally advanced esophageal squamous cell carcinoma. Cancer Biol Ther 2024; 25:2371632. [PMID: 38946404 PMCID: PMC11218796 DOI: 10.1080/15384047.2024.2371632] [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: 12/15/2022] [Accepted: 06/19/2024] [Indexed: 07/02/2024] Open
Abstract
To investigate the impact of the effective radiation dose to immune cells (EDIC) and gross tumor volume (GTV) on lymphopenia and survival in patients with locally advanced esophageal squamous cell carcinoma (LAESCC). Between January 2013 and December 2020, 272 LAESCC patients were treated with definitive radiotherapy in two institutions. Based on radiation doses to the lungs, heart, and body region scanned, EDIC was calculated as an equal uniform dose to the total blood considering blood flow and fraction effect. The radiotherapy plan was used to calculate the GTVs. Lymphopenia was graded based on the lowest lymphocyte count during RT. The overall survival (OS), progress-free survival (PFS), and local recurrence-free survival (LRFS) were analyzed statistically. The lowest lymphocyte count was significantly correlated with EDIC (r= -0.389, p < .001) and GTV (r= -0.211, p < .001). Lymphopenia, EDIC, and GTV are risk factors for patients with ESCC. In a Kaplan-Meier analysis with EDIC and GTV as stratification factors, lymphopenia was not associated with OS in the EDIC>12.9 Gy group (p = .294)and EDIC ≤ 12.9 Gy group, and it was also not associated with OS in GTV>68.8 cm3 group (p = .242) and GTV ≤ 68.8 cm3 group(p = .165). GTV and EDIC had an impact on the relationship between lymphopenia and OS in patients with LAESCC undergoing definitive RT. Poorer OS, PFS, and LRFS are correlated with lymphopenia, higher EDIC, and larger GTV.
Collapse
Affiliation(s)
- Danjing Luo
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiulu Zhong
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Haiying Yue
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jue Wang
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qianfu Liang
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wenqi Liu
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaodong Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
- Department of Oncology, Wuming Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
9
|
Song KW, Scott BJ. CAR T-cell therapy for gliomas. Curr Opin Neurol 2024; 37:672-681. [PMID: 39498846 DOI: 10.1097/wco.0000000000001318] [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: 11/07/2024]
Abstract
PURPOSE OF REVIEW To review the landscape of chimeric antigen receptor T-cell (CAR T) therapy for gliomas as seen in recently published trials and discuss on-going challenges with new cancer immunotherapy treatments. RECENT FINDINGS Given how CAR T therapy has revolutionized the treatment of several hematologic malignancies, there has been increasing interest in using immunotherapy, and particularly CAR T therapy for gliomas. Within the past decade, several first in human trials have published early patient experiences showing treatment is generally well tolerated but with limited efficacy, which may be improving with newer evolutions in CAR T design to overcome known resistance mechanisms in glioma treatment. SUMMARY CAR T therapy is a promising avenue of treatment for high-grade gliomas, which have a universally poor prognosis as well as limited therapeutics. There are a growing number of CAR T clinical trials for CNS tumors and thus, an understanding of their treatment strategies, toxicity management, and overcoming resistance mechanisms will be important for both clinical practice and to identify areas for future research.
Collapse
Affiliation(s)
- Kun-Wei Song
- Department of Neurology, Stanford University School of Medicine
- Stanford Neuro-Immuno-Oncology (NIO) Program, Stanford, California, USA
| | - Brian J Scott
- Department of Neurology, Stanford University School of Medicine
- Stanford Neuro-Immuno-Oncology (NIO) Program, Stanford, California, USA
| |
Collapse
|
10
|
Koukourakis IM, Georgakopoulos I, Desse D, Tiniakos D, Kouloulias V, Zygogianni A. Lymphopenia is an adverse prognostic factor in rectal adenocarcinoma patients receiving long-course chemoradiotherapy. Radiat Oncol J 2024; 42:263-272. [PMID: 39748527 DOI: 10.3857/roj.2024.00052] [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: 01/17/2024] [Accepted: 03/08/2024] [Indexed: 01/04/2025] Open
Abstract
PURPOSE Neoadjuvant radiotherapy (RT) or chemoradiotherapy (CRT) is the standard treatment for locally advanced rectal adenocarcinoma. The recent emerging data on preoperative immunotherapy as an effective therapeutic modality for mismatch repair deficient rectal carcinomas suggests that the immune system plays a significant role in tumor eradication. Although RT has been shown to stimulate anti-tumor immunity, it also leads to substantial lymphopenia, hindering the effect of immune response. MATERIALS AND METHODS We retrospectively analyzed 33 rectal adenocarcinoma patients who underwent CRT in our department, aiming to identify the effects of CRT on the peripheral blood lymphocyte counts (LC) and the potential impact of CRT-induced lymphopenia on tumor response and prognosis of patients. RESULTS A statistically significant decrease in the LC of patients was observed after CRT (median values of 2,184/μL and 517/μL before and after treatment, respectively; p < 0.001). While no correlation between ypT-stage, ypN status, and LC was found, poor tumor regression grade was significantly associated with lower LC (p = 0.036). Moreover, lymphopenia was associated with poorer distant metastasis-free survival (p = 0.003). Distant metastases were documented in 0% of patients with post-CRT LC above 518/μL vs. 44.5% of patients with lower LC values. CONCLUSION Although further investigation is demanded, given the limited number of patients analyzed in the study, lymphopenia emerges as a significant adverse event that rectal adenocarcinoma patients face during treatment with neoadjuvant CRT, with subsequent implications on tumor response and prognosis. Protection of the immune system during CRT emerges as an important target for clinical research.
Collapse
Affiliation(s)
- Ioannis M Koukourakis
- Radiation Oncology Unit, 1st Department of Radiology, Medical School, Aretaieion Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Georgakopoulos
- Radiation Oncology Unit, 1st Department of Radiology, Medical School, Aretaieion Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitra Desse
- Radiation Oncology Unit, 1st Department of Radiology, Medical School, Aretaieion Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dina Tiniakos
- Department of Pathology, Aretaieion University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Vassilios Kouloulias
- Radiation Oncology Unit, 2nd Department of Radiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Zygogianni
- Radiation Oncology Unit, 1st Department of Radiology, Medical School, Aretaieion Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
11
|
Pham TN, Coupey J, Rousseau M, Thariat J, Valable S. Revealing the effect of X-ray or proton brain irradiation on systemic inflammation and leukocyte subpopulation interplay in rodents. J Leukoc Biol 2024; 116:1530-1543. [PMID: 38952292 DOI: 10.1093/jleuko/qiae156] [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: 02/15/2024] [Revised: 05/29/2024] [Accepted: 07/01/2024] [Indexed: 07/03/2024] Open
Abstract
The absolute lymphocyte count (ALC), lymphocyte-to-monocyte ratio (LMR), and neutrophil-to-lymphocyte ratio (NLR) offer convenient means to assess systemic inflammation post-cancer treatment, which influences treatment outcomes. Understanding these biomarker variations and leukocyte subpopulation interplay is crucial for optimizing radiotherapy. Herein, leukocyte subpopulations (T-CD4+, T-CD8+, B cells, NK cells, neutrophils, monocytes) during and after brain irradiation (using X-rays or protons) in tumor-free mice were used to compute ALC, LMR, and NLR, on which radiation parameter influence was assessed by principal component analysis (PCA). NLR kinetics was further examined using modeling. Leukocyte subpopulation interplays and their response to radiation parameters were examined using PCA and correlation analysis. Under X-rays, ALC and LMR decreased, with ALC recovered to baseline after irradiation, but not LMR. Both X-rays and protons increased the NLR during irradiation, recovering in protons but not X-rays. Both irradiation volume and dose rate had a pronounced effect on the NLR. Leukocyte subpopulation interplay was observed under X-rays and protons, normalizing in the proton group by day 28. Lymphopenia was observed in all lymphocyte subpopulations under X-ray irradiation but not protons. The recovery patterns varied among the subpopulations. Neutrophil counts increased during irradiation, with the recovery of protons, but not X-rays, by day 28. Interplays between NK cells and myeloid subpopulations were evident under X-rays but not protons. Importantly, no interplay was detected between myeloid cells and T/B cells, indicating that LMR and NLR variations were primarily due to independent responses to brain irradiation. A tumor-free experimental mouse model was used to study the effects of brain radiotherapy on systemic immunity. When administering fractionated irradiation with a total dose of 20 Gy using a vertical beam to either the whole brain or hemi-brain, proton irradiation had fewer adverse impacts on the immune system compared to X-rays in tumor-free rodents.
Collapse
Affiliation(s)
- Thao-Nguyen Pham
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, Normandy, France
- Laboratoire de physique corpusculaire UMR6534 IN2P3/ENSICAEN, France-Normandie Université, 14000 Caen, Normandy, France
| | - Julie Coupey
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, Normandy, France
| | - Marc Rousseau
- Laboratoire de physique corpusculaire UMR6534 IN2P3/ENSICAEN, France-Normandie Université, 14000 Caen, Normandy, France
| | - Juliette Thariat
- Laboratoire de physique corpusculaire UMR6534 IN2P3/ENSICAEN, France-Normandie Université, 14000 Caen, Normandy, France
- Department of Radiation Oncology, Centre François Baclesse, 14000 Caen, Normandy, France
| | - Samuel Valable
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, Normandy, France
| |
Collapse
|
12
|
Wang M, Liu G, Liang Y, Lyu Z, Tang Z, Tan F, Wei R. Clinical results of helical tomotherapy for high-grade gliomas. Int J Radiat Biol 2024; 100:1683-1695. [PMID: 39495095 DOI: 10.1080/09553002.2024.2418500] [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: 08/01/2023] [Revised: 08/13/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Radiotherapy-related damage of normal tissue inevitably influences the treatment outcomes in the context of high-grade gliomas (HGGs) treatment. We reported the survival outcomes and toxicities of patients with HGG treated with helical tomotherapy (HT) and the prognostic factors were analyzed. MATERIALS AND METHODS A total of 67 patients (29 had grade III and 38 had grade IV HGGs) who received HT between January 2016 and June 2020 were analyzed. Overall survival (OS) and progression-free survival (PFS) from the beginning of HT and OS from surgery were assessed, and toxicity and disease control were described briefly. RESULTS For patients with grade III HGGs, median OS (mOS) and median PFS (mPFS) from the beginning of HT were 68.933 and 62.967 months, respectively. For patients with grade IV HGGs, mOS and mPFS from the beginning of HT were 19.667 and 7.23 months, respectively. No grade ≥3 acute or late nonhematologic toxicities were observed. Multivariable Cox regression analysis showed that methylguanine methyltransferase (MGMT) methylated status, age, number of lesions, WHO grade, and monocyte count for PFS were significant. Age, monocyte count, and isocitrate dehydrogenase (IDH) status for OS. CONCLUSION Treatment of HGGs with HT appears to be potentially effective and safe. HT is promising for glioblastomas (GBM), especially complex cases with infratentorial involvement or multiple lesions. This study highlighted the potential clinical significance of systemic inflammation indicators in predicting survival and disease progression.
Collapse
Affiliation(s)
- Min Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Gui Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Liang
- Department of Oncology, Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Zhiping Lyu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziqing Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fang Tan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rui Wei
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
Song YC, Chen SY, Zhao XR, Jing H, Fang H, Tang Y, Hu SY, Song YW, Jin J, Liu YP, Qi SN, Sun GY, Zhong QZ, Du XH, Liu J, Li YX, Wang SL. Prognostic value of lymphocytes in patients with breast cancer receiving radiotherapy after breast-conserving surgery: A post hoc analysis of a phase III randomized trial. Radiother Oncol 2024; 199:110390. [PMID: 38897316 DOI: 10.1016/j.radonc.2024.110390] [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: 02/28/2024] [Revised: 06/11/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE To evaluate the prognostic value of peripheral lymphocyte count (PLC) in the breast cancer patients after breast-conserving surgery (BCS) with radiotherapy (RT). METHODS AND MATERIALS This post hoc analysis was performed using data of 628 patients from a phase III, randomized controlled trial comparing hypofractionated RT (HFRT) with conventional fractionated RT (CFRT) after BCS. PLCs were obtained before, during, and after RT until the 1-year follow-up. The optimal cut-off PLCs were determined using the maxstat package in R. Survival rates were estimated using the Kaplan-Meier method and compared with the log-rank test. RESULTS A total of 275 (46.1 %) patients developed lymphopenia during RT, among them, 17 (2.8 %) had grade 3 lymphopenia and no one developed grade 4 lymphopenia. With a median follow-up of 110.8 months, patients with pre-RT PLCs of < 1.77 × 109/L had a significantly lower 10-year breast cancer-specific survival (BCSS) rate (P = 0.013) and overall survival (OS) rate (P = 0.026). Patients with a nadir PLC of < 1.35 × 109/L had a significantly poorer 10-year OS rate (P = 0.048). Multivariate analysis showed that a pre-RT PLC of < 1.77 × 109/L was an independent factor influencing BCSS and OS, while the effect of the nadir PLC did not remain significant. Neither PLC nor lymphopenia recovery at post-RT 1, 3, and 6 months and 1 year was associated with survival. CONCLUSIONS Radiation-induced lymphopenia in patients with breast cancer after BCS tends to be mild. The lower pre-RT PLC predicted poorer survival.
Collapse
Affiliation(s)
- Yu-Chun 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
| | - Si-Ye 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - Qiu-Zi Zhong
- Department of Radiation Oncology, Beijing Hospital, Ministry of Health, Beijing, China
| | - Xiang-Hui Du
- Department of Radiation Therapy, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Juan Liu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, 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.
| | - 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
|
14
|
Xia F, Wang Y, Wang H, Shen L, Xiang Z, Zhao Y, Zhang H, Wan J, Zhang H, Wang Y, Wu R, Wang J, Yang W, Zhou M, Zhou S, Chen Y, Zhang Z, Wu X, Xuan Y, Wang R, Sun Y, Tong T, Zhang X, Wang L, Huang D, Sheng W, Yan H, Yang X, Shen Y, Xu Y, Zhao R, Mo M, Cai G, Cai S, Xu Y, Zhang Z. Randomized Phase II Trial of Immunotherapy-Based Total Neoadjuvant Therapy for Proficient Mismatch Repair or Microsatellite Stable Locally Advanced Rectal Cancer (TORCH). J Clin Oncol 2024; 42:3308-3318. [PMID: 38950321 DOI: 10.1200/jco.23.02261] [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: 10/17/2023] [Revised: 01/17/2024] [Accepted: 04/17/2024] [Indexed: 07/03/2024] Open
Abstract
PURPOSETo assess whether the integration of PD-1 inhibitor with total neoadjuvant therapy (iTNT) can lead to an improvement in complete responses (CRs) and favors a watch-and-wait (WW) strategy in patients with proficient mismatch repair or microsatellite stable (pMMR/MSS) locally advanced rectal cancer (LARC).PATIENTS AND METHODSWe conducted a prospective, multicenter, randomized, open-label, phase II trial using a pick-the-winner design. Eligible patients with clinical T3-4 and/or N+ rectal adenocarcinoma were randomly assigned to group A for short-course radiotherapy (SCRT) followed by six cycles of consolidation immunochemotherapy with capecitabine and oxaliplatin and toripalimab or to group B for two cycles of induction immunochemotherapy followed by SCRT and the rest four doses. Either total mesorectal excision or WW was applied on the basis of tumor response. The primary end point was CR which included pathological CR (pCR) after surgery and clinical CR (cCR) if WW was applicable, with hypothesis of an increased CR of 40% after iTNT compared with historical data of 25% after conventional TNT.RESULTSOf the 130 patients enrolled, 121 pMMR/MSS patients were evaluable (62 in group A and 59 in group B). At a median follow-up of 19 months, CR was achieved at 56.5% in group A and 54.2% in group B. Both groups fulfilled the predefined statistical hypothesis (P < .001). Both groups reported a pCR rate of 50%. Respectively, 15 patients in each group underwent WW and remained disease free. The most frequent grade 3 to 4 toxicities were thrombocytopenia and neutropenia. Patients in group A had higher rate of cCR (43.5% v 35.6%) at restaging and lower rate of grade 3 to 4 thrombocytopenia (24.2% v 33.9%) during neoadjuvant treatment.CONCLUSIONThe iTNT regimens remarkably improved CR rates in pMMR/MSS LARC compared with historical benchmark with acceptable toxicity. Up-front SCRT followed by immunochemotherapy was selected for future definitive study.
Collapse
Affiliation(s)
- Fan Xia
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yaqi Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Wang
- Department of Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Lijun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zuolin Xiang
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yutian Zhao
- Department of Radiation Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Huojun Zhang
- Department of Radiation Oncology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Juefeng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruiyan Wu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingwen Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wang Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Menglong Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shujuan Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yajie Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xian Wu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Xuan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Renjie Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yiqun Sun
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Tong Tong
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xun Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lei Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Dan Huang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Weiqi Sheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Hao Yan
- Department of Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Xu Yang
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuxin Shen
- Department of Radiation Oncology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yu Xu
- Department of Radiation Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ruping Zhao
- Department of Radiation Oncology, Jia Hui International Hospital, Shanghai, China
- Department of Radiotherapy, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Miao Mo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Cancer Prevention, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Guoxiang Cai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Sanjun Cai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ye Xu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Yang G, Koom WS, Lee BM, Isozaki T, Shinoto M, Yamada S, Seong J. Reduced Risk of Severe Radiation-Induced Lymphopenia in Carbon Ion Radiation Therapy for Locally Advanced Pancreatic Cancer: A Comparative Analysis of Carbon Versus Photon Therapy. Int J Radiat Oncol Biol Phys 2024; 120:544-554. [PMID: 38713122 DOI: 10.1016/j.ijrobp.2024.04.003] [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: 09/25/2023] [Revised: 03/15/2024] [Accepted: 04/03/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE Radiation-induced lymphopenia (RIL) is associated with poor prognosis in patients with locally advanced pancreatic cancers. However, there are no reports comparing the effects of carbon ion radiation therapy (CIRT) and photon beam radiation therapy (RT) on the development of RIL. Differences in RIL after CIRT or photon beam RT and predictive factors for RIL in patients with locally advanced pancreatic cancer were investigated. MATERIALS AND METHODS This retrospective study cohort included 834 patients who received concurrent chemoradiotherapy (CCRT) in 2 separate institutions: 337 and 497 in the CIRT and photon beam RT groups, respectively. Severe RIL was defined as an absolute lymphocyte count (ALC) <0.5 × 109 cells/L. A 1:1 propensity score-matching analysis was performed between the CIRT and photon beam RT groups. Patients were categorized into 3 groups according to the development of recovery from severe RIL: no severe RIL (Group A), recovery from severe RIL (Group B), and no recovery from severe RIL (Group C). Logistic regression analysis was performed to identify the predictive value of severe RIL. The prognostic factors of overall survival (OS) were determined using Cox regression analysis. RESULTS After propensity score matching, the baseline ALC and planning target volume of the CIRT and photon beam RT groups were comparable. During CCRT, the ALC of the entire cohort decreased and was significantly lower in the photon beam RT group than in the CIRT group (P < .001). Multivariate logistic regression analysis showed that CIRT reduced severe RIL more than photon beam RT. After adjusting for other factors, the RT modality and RIL were significantly associated with OS. Photon beam RT showed a significantly worse OS than CIRT, and Group C showed a significantly worse OS than Group A. CONCLUSIONS CIRT seems to reduce the development of severe RIL. The RT modality and development/recovery from severe RIL were associated with OS in patients who received CCRT for locally advanced pancreatic cancer. The reduction of severe RIL through optimized RT may be essential for improving treatment outcomes.
Collapse
Affiliation(s)
- Gowoon Yang
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byung Min Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tetsuro Isozaki
- QST Hospital, National Institutes for Quantum Science and Technology, Inageku, Chiba, Japan
| | - Makoto Shinoto
- QST Hospital, National Institutes for Quantum Science and Technology, Inageku, Chiba, Japan
| | - Shigeru Yamada
- QST Hospital, National Institutes for Quantum Science and Technology, Inageku, Chiba, Japan.
| | - Jinsil Seong
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Tubin S, Ashdown ML, Ahmed MM, Guha C, Salerno G, Celedin B, Trummer B, Demschar S, Raunik W. Novel time-synchronized immune-guided partial tumor irradiation: Proof of principle trial. Radiother Oncol 2024; 199:110442. [PMID: 39069088 DOI: 10.1016/j.radonc.2024.110442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND AND PURPOSE Radiotherapy for bulky tumors often results in palliation with suboptimal outcomes. The prognosis is worsened by immunosuppression caused by radio-chemotherapy, negatively impacting on survival. Novel Partial Tumor Irradiation (PTI) was designed to spare the Peritumoral Immune Microenvironment (PIM) and to be delivered synchronously with immune activity peaks, thus enhancing both local and distant tumor control through immunostimulation. MATERIALS AND METHODS Present proof-of-principle trial enrolled 26 patients with bulky tumors, comparing outcomes between treatments administered at immune activity peaks versus troughs. The primary endpoint was local-bystander and distal-abscopal response-rate. Secondary endpoints included overall-, progression-free-, cancer-specific survival, neoadjuvant and immunomodulatory potential. RESULTS All measured outcomes were significantly influenced by treatment-timing. The bystander and abscopal response rates were 77% and 41%, respectively. PTI significantly upregulated pro-inflammatory and cell-death-inducing pathways improving the efficacy of radiotherapy by highly complex tumors. CONCLUSIONS This study highlights the profound impact PTI can have on a highly palliative patient cohort previously deemed beyond therapeutic hope. With 41 % of these patients still alive after a median follow-up of 50 months, PTI offers a potential lifeline for those facing advanced, treatment-resistant cancers. This approach generated also distant immunogenic anti-tumor responses, offering a promising new avenue for the treatment of advanced cancers.
Collapse
Affiliation(s)
- S Tubin
- Medaustron Center for Ion Therapy, Marie-Curie Strasse 5, Wiener Neustadt 2700, Austria; KABEG Klinikum Klagenfurt, Institute of Radiation Oncology, Feschnigstraße 11 9020, Klagenfurt am Wörthersee, Austria; Heidelberg University Hospital, Department of Radiation Oncology and Radiation Therapy, Im Neuenheimer Feld 400 69120, Heidelberg, Germany; Division of Radiation Biology and Molecular Therapeutics at the Department of Radiation Oncology, Albert Einstein College of Medicine, 111 E. 210th Street Klau 3 Bronx, NY 10467, New York, United States.
| | - M L Ashdown
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, 3010, Melbourne, Australia
| | - M M Ahmed
- Division of Radiation Biology and Molecular Therapeutics at the Department of Radiation Oncology, Albert Einstein College of Medicine, 111 E. 210th Street Klau 3 Bronx, NY 10467, New York, United States
| | - C Guha
- Montefiore Medical Center Radiation Oncology, 111 E 210th St, New York, NY, United States
| | - G Salerno
- Department of Neurosciences, Mental Health and Sensory Organs / Department of Clinical and Molecular Medicine, Universita' La Sapienza Roma, Ospedale Sant' Andrea, Via di Grottarossa, 1035 00189, Rome, RM, Italy.
| | - B Celedin
- KABEG Klinikum Klagenfurt, Institute of Radiation Oncology, Feschnigstraße 11 9020, Klagenfurt am Wörthersee, Austria
| | - B Trummer
- Center for Interdisciplinary Pain Therapy, Oncology and Palliative Care, Klinikum Klagenfurt am Wörthersee, Feschnigstr. 11 9020, Klagenfurt am Wörthersee, Austria
| | - S Demschar
- Center for Interdisciplinary Pain Therapy, Oncology and Palliative Care, Klinikum Klagenfurt am Wörthersee, Feschnigstr. 11 9020, Klagenfurt am Wörthersee, Austria
| | - W Raunik
- KABEG Klinikum Klagenfurt, Institute of Radiation Oncology, Feschnigstraße 11 9020, Klagenfurt am Wörthersee, Austria
| |
Collapse
|
17
|
Hong W, Zhang L, Qi Y, Wang Y, Wang W. Impact of Chemotherapy on Circulating Lymphocyte Subsets in Lung Cancer Patients. Cancer Manag Res 2024; 16:1205-1213. [PMID: 39282611 PMCID: PMC11401525 DOI: 10.2147/cmar.s475967] [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: 05/18/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose Lung cancer remains a leading cause of cancer-related death and chemotherapy stands as a fundamental component in therapy. Chemotherapy-induced myelosuppression encompasses a spectrum of hematological declines, including not only neutrophils but also lymphocytes, hemoglobin levels and platelets. This retrospective cohort study investigates alterations in peripheral blood lymphocyte subsets. By uncovering these changes, our goal is to refine patient management strategies, ensuring that the benefits of chemotherapy are maximized while minimizing its detrimental effects. Patients and Methods We retrospectively analyzed 159 lung cancer patients. Patients were categorized as "NT" (n=108, no previous anti-tumor therapy), and "PT" (n=51, prior therapy followed by at least a two-month treatment-free interval). Post-chemotherapy, patients were reassessed and grouped into "EarlyCycle" for those who underwent four or fewer cycles, and "LateCycle" for those who underwent more than four cycles. Results The study focused on analyzing the percentages of lymphocyte subsets, including T cells (CD4+, CD8+), B cells, and natural killer (NK) cells, across these groups. For T cells, the EarlyCycle group exhibited a significant increase compared to NT (0.7783 vs 0.7271; p=0.0017) and PT (0.7783 vs 0.6804; p=1.6e-05). B cells showed a significant decrease from NT to LateCycle (0.1014 vs 0.0817; p=2.2e-05) and from PT to LateCycle (0.1317 vs 0.0817; p=6.2e-10). NK cells significantly decreased in the EarlyCycle group compared to NT (0.1109 vs 0.1462; p=0.00816) and PT (0.1109 vs 0.1513; p=0.00992), with no significant change in the LateCycle group compared to either NT or PT (p>0.05). Conclusion Chemotherapy significantly affects lymphocyte subsets in a treatment-specific manner. The EarlyCycle group experienced a reduction in NK cell and an increase in T cell, suggesting a damage of innate immunity and an early shift towards adaptive immunity. The LateCycle group showed a substantial decrease in B cell, indicating a delayed effect on humoral immunity components.
Collapse
Affiliation(s)
- Wei Hong
- Oncology, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| | - Lei Zhang
- Oncology, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| | - Youkun Qi
- Pharmacy, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| | - Yanjun Wang
- Oncology, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| | - Wentao Wang
- Critical Care Medicine, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| |
Collapse
|
18
|
Colen J, Nguyen C, Liyanage SW, Aliotta E, Chen J, Alonso C, Romano K, Peach S, Showalter T, Read P, Larner J, Wijesooriya K. Predicting radiation-induced immune suppression in lung cancer patients treated with stereotactic body radiation therapy. Med Phys 2024; 51:6485-6500. [PMID: 38837261 PMCID: PMC11489021 DOI: 10.1002/mp.17181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/14/2024] [Accepted: 04/21/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Stereotactic body radiation therapy (SBRT) is known to modulate the immune system and contribute to the generation of anti-tumor T cells and stimulate T cell infiltration into tumors. Radiation-induced immune suppression (RIIS) is a side effect of radiation therapy that can decrease immunological function by killing naive T cells as well as SBRT-induced newly created effector T cells, suppressing the immune response to tumors and increasing susceptibility to infections. PURPOSE RIIS varies substantially among patients and it is currently unclear what drives this variability. Models that can accurately predict RIIS in near real time based on treatment plan characteristics would allow treatment planners to maintain current protocol specific dosimetric criteria while minimizing immune suppression. In this paper, we present an algorithm to predict RIIS based on a model of circulating blood using early stage lung cancer patients treated with SBRT. METHODS This Python-based algorithm uses DICOM data for radiation therapy treatment plans, dose maps, patient CT data sets, and organ delineations to stochastically simulate blood flow and predict the doses absorbed by circulating lymphocytes. These absorbed doses are used to predict the fraction of lymphocytes killed by a given treatment plan. Finally, the time dependence of absolute lymphocyte count (ALC) following SBRT is modeled using longitudinal blood data up to a year after treatment. This model was developed and evaluated on a cohort of 64 patients with 10-fold cross validation. RESULTS Our algorithm predicted post-treatment ALC with an average error of0.24 ± 0.21 × 10 9 $0.24 \pm 0.21 \times {10}^9$ cells/L with 89% of the patients having a prediction error below 0.5 × 109 cells/L. The accuracy was consistent across a wide range of clinical and treatment variables. Our model is able to predict post-treatment ALC < 0.8 (grade 2 lymphopenia), with a sensitivity of 81% and a specificity of 98%. This model has a ∼38-s end-to-end prediction time of post treatment ALC. CONCLUSION Our model performed well in predicting RIIS in patients treated using lung SBRT. With near-real time model prediction time, it has the capability to be interfaced with treatment planning systems to prospectively reduce immune cell toxicity while maintaining national SBRT conformity and plan quality criteria.
Collapse
Affiliation(s)
- Jonathan Colen
- University of Virginia, Department of Physics,
Charlottesville, Virginia, USA
- Old Dominion University, Joint Institute on Advanced
Computing for Environmental Studies, Norfolk, Virginia, USA
- Hampton Roads Biomedical Research Consortium, Portsmouth,
Virginia, USA
| | - Cam Nguyen
- University of Virginia, Department of Physics,
Charlottesville, Virginia, USA
| | - Seth W. Liyanage
- Stanford University, Department of Mechanical Engineering,
Stanford, California, USA
| | - Eric Aliotta
- University of Virginia, Department of Radiation Oncology,
Charlottesville, Virginia, USA
| | - Joe Chen
- University of Virginia, Department of Radiation Oncology,
Charlottesville, Virginia, USA
| | - Clayton Alonso
- University of Virginia, Department of Radiation Oncology,
Charlottesville, Virginia, USA
| | - Kara Romano
- University of Virginia, Department of Radiation Oncology,
Charlottesville, Virginia, USA
| | - Sean Peach
- University of Virginia, Department of Radiation Oncology,
Charlottesville, Virginia, USA
| | - Timothy Showalter
- University of Virginia, Department of Radiation Oncology,
Charlottesville, Virginia, USA
| | - Paul Read
- University of Virginia, Department of Radiation Oncology,
Charlottesville, Virginia, USA
| | - James Larner
- University of Virginia, Department of Radiation Oncology,
Charlottesville, Virginia, USA
| | - Krishni Wijesooriya
- University of Virginia, Department of Physics,
Charlottesville, Virginia, USA
- University of Virginia, Department of Radiation Oncology,
Charlottesville, Virginia, USA
| |
Collapse
|
19
|
Hu Z, Mohan R, Chu Y, Wang X, van Rossum PS, Chen Y, Grayson ME, Gearhardt AG, Grassberger C, Zhi D, Hobbs BP, Lin SH, Cao W. Clinical Translation of a Deep Learning Model of Radiation-Induced Lymphopenia for Esophageal Cancer. Int J Part Ther 2024; 13:100624. [PMID: 39228692 PMCID: PMC11369390 DOI: 10.1016/j.ijpt.2024.100624] [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: 06/26/2024] [Accepted: 07/29/2024] [Indexed: 09/05/2024] Open
Abstract
Purpose Radiation-induced lymphopenia is a common immune toxicity that adversely impacts treatment outcomes. We report here our approach to translate a deep-learning (DL) model developed to predict severe lymphopenia risk among esophageal cancer into a strategy for incorporating the immune system as an organ-at-risk (iOAR) to mitigate the risk. Materials and Methods We conducted "virtual clinical trials" utilizing retrospective data for 10 intensity-modulated radiation therapy (IMRT) and 10 passively-scattered proton therapy (PSPT) esophageal cancer patients. For each patient, additional treatment plans of the modality other than the original were created employing standard-of-care (SOC) dose constraints. Predicted values of absolute lymphocyte count (ALC) nadir for all plans were estimated using a previously-developed DL model. The model also yielded the relative magnitudes of contributions of iOARs dosimetric factors to ALC nadir, which were used to compute iOARs dose-volume constraints, which were incorporated into optimization criteria to produce "IMRT-enhanced" and "intensity-modulated proton therapy (IMPT)-enhanced" plans. Results Model-predicted ALC nadir for the original IMRT (IMRT-SOC) and PSPT plans agreed well with actual values. IMPT-SOC showed greater immune sparing vs IMRT and PSPT. The average mean body doses were 13.10 Gy vs 7.62 Gy for IMRT-SOC vs IMPT-SOC for patients treated with IMRT-SOC; and 8.08 Gy vs 6.68 Gy for PSPT vs IMPT-SOC for patients treated with PSPT. For IMRT patients, the average predicted ALC nadir of IMRT-SOC, IMRT-enhanced, IMPT-SOC, and IMPT-enhanced was 281, 327, 351, and 392 cells/µL, respectively. For PSPT patients, the average predicted ALC nadir of PSPT, IMPT-SOC, and IMPT-enhanced was 258, 316, and 350 cells/µL, respectively. Enhanced plans achieved higher predicted ALC nadir, with an average improvement of 40.8 cells/µL (20.6%). Conclusion The proposed DL model-guided strategy to incorporate the immune system as iOAR in IMRT and IMPT optimization has the potential for radiation-induced lymphopenia mitigation. A prospective clinical trial is planned.
Collapse
Affiliation(s)
- Zongsheng Hu
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Radhe Mohan
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yan Chu
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xiaochun Wang
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Yiqing Chen
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Madison E. Grayson
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Angela G. Gearhardt
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Clemens Grassberger
- Department of Radiation Oncology, University of Washington, Seattle, Washington, USA
| | - Degui Zhi
- The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Brian P. Hobbs
- Department of Population Health, The University of Texas at Austin, Austin, Texas, USA
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wenhua Cao
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
20
|
Takeda K, Umezawa R, Yamamoto T, Takahashi N, Suzuki Y, Kishida K, Omata S, Jingu K. Lymphopenia after palliative radiotherapy for vertebral metastases. JOURNAL OF RADIATION RESEARCH 2024; 65:523-531. [PMID: 38818633 PMCID: PMC11262856 DOI: 10.1093/jrr/rrae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/04/2024] [Indexed: 06/01/2024]
Abstract
Lymphopenia is a well-known side effect of radiotherapy and has been shown to have a negative impact on patient outcomes. However, the extent of lymphopenia caused by palliative radiotherapy and its effect on patient prognosis has not been clarified. The aim of this study was to determine the incidence and severity of lymphopenia after palliative radiotherapy for vertebral metastases and to determine their effects on patients' survival outcomes. We conducted a retrospective analysis for patients who underwent palliative radiotherapy for vertebral metastases and could be followed up for 12 weeks. Lymphocyte counts were documented at baseline and throughout the 12-week period following the start of radiotherapy and their medians and interquartile ranges (IQRs) were recorded. Exploratory analyses were performed to identify predictive factors for lymphopenia and its impact on overall survival (OS). A total of 282 cases that met the inclusion criteria were analyzed. The median baseline lymphocyte count was 1.26 × 103/μl (IQR: 0.89-1.72 × 103/μl). Peak lymphopenia occurred at a median of 26 days (IQR: 15-45 days) with a median nadir of 0.52 × 103/μl (IQR: 0.31-0.81 × 103/μl). Long-term analysis of patients surviving for 1 year showed that lymphopenia persisted at 1 year after radiotherapy. The main irradiation site, radiation field length and pretreatment lymphocyte count were significantly related to grade 3 or higher lymphopenia. Lymphopenia was identified as a significant predictor of OS by multivariate Cox regression analysis. This study demonstrated the incidence of lymphopenia after palliative radiotherapy for vertebral metastases and its effect on patients' OS.
Collapse
Affiliation(s)
- Kazuya Takeda
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi Japan
- Department of Radiation Oncology, South Miyagi Medical Center, 38-1 Nishi, Ogawara, Shibata 989-1253, Miyagi, Japan
| | - Rei Umezawa
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi Japan
| | - Takaya Yamamoto
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi Japan
| | - Noriyoshi Takahashi
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi Japan
| | - Yu Suzuki
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi Japan
| | - Keita Kishida
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi Japan
| | - So Omata
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi Japan
| | - Keiichi Jingu
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi Japan
| |
Collapse
|
21
|
Coupey J, Leblond MM, Hue ES, Valable S. Flow cytometry detection and quantification of circulating leukocyte subpopulations in mice after brain irradiation. Methods Cell Biol 2024; 189:135-152. [PMID: 39393880 DOI: 10.1016/bs.mcb.2024.06.004] [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: 10/13/2024]
Abstract
In the context of high-grade gliomas such as glioblastoma (GBM), the immune part of the tumor microenvironment (TME) is involved in tumor growth and tumor recurrence. It is mostly represented by high amount of macrophages and low amount of lymphocytes. GBM in itself as well as x-ray-based radiotherapy, a standard treatment for brain tumors, are also associated with systemic effects like lymphopenia that correlates with a poor prognosis. This contributes to the immune-suppressive nature of the TME and may explain the lack of the anti-tumor immune response. Radiation-induced lymphopenia (RIL) is generally evaluated on CD4+ and CD8+ count or on a CBC (complete blood count), but the heterogeneity of the subtypes prompts us to explore them in detail to better understand the cellular response to brain irradiation. To facilitate and develop the evaluation of x-ray brain exposure on circulating immune cells, we developed a reproducible and reliable method to quantify the variation of lymphoid and myeloid subtypes using flow cytometry after brain irradiation in the rodent.
Collapse
Affiliation(s)
- Julie Coupey
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP Cyceron, Caen, France
| | - Marine M Leblond
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Erika S Hue
- LABÉO, Saint-Contest, France; Normandie Université, UNICAEN BIOTARGEN, Saint-Contest, France
| | - Samuel Valable
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP Cyceron, Caen, France.
| |
Collapse
|
22
|
Kim N, Lee J, Shin H, Shin J, Nam DH, Lee JI, Seol HJ, Kong DS, Choi JW, Chong K, Lee WJ, Chang JH, Kang SG, Moon JH, Cho J, Lim DH, Yoon HI. Nomogram for radiation-induced lymphopenia in patients receiving intensity-modulated radiotherapy based-chemoradiation therapy for newly diagnosed glioblastoma: A multi-institutional study. Clin Transl Radiat Oncol 2024; 47:100799. [PMID: 38884005 PMCID: PMC11176633 DOI: 10.1016/j.ctro.2024.100799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/09/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
Purpose Severe lymphopenia (SLP) has emerged as a significant prognostic factor in glioblastoma. Intensity-modulated radiation therapy (IMRT)-based radiation therapy (RT) is suggested to minimize the risk of SLP. This study aimed to evaluate SLP incidence based on multi-institutional database in patients with GBM treated with IMRT and develop a predictive nomogram. Patients and methods This retrospective study reviewed data from 348 patients treated with IMRT-based concurrent chemoradiation therapy (CCRT) at two major hospitals from 2016 to 2021. After multivariate regression analysis, a nomogram was developed and internally validated to predict SLP risk. Results During treatment course, 21.0% of patients developed SLP and SLP was associated with poor overall survival outcomes in patients with GBM. A newly developed nomogram, incorporating gender, pre-CCRT absolute lymphocyte count, and brain mean dose, demonstrated fair predictive accuracy (AUC 0.723). Conclusions This study provides the first nomogram for predicting SLP in patients with GBM treated with IMRT-based CCRT, with acceptable predictive accuracy. The findings underscore the need for dose optimization and radiation planning to minimize SLP risk. Further external validation is crucial for adopting this nomogram in clinical practice.
Collapse
Affiliation(s)
- Nalee Kim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Joongyo Lee
- Department of Radiation Oncology, Gangnam Severance Hospital, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyunju Shin
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Jungwook Shin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, United States
| | - Do-Hyun Nam
- Department of Neurosurgery, Brain Tumor Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Jung-Il Lee
- Department of Neurosurgery, Brain Tumor Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Ho Jun Seol
- Department of Neurosurgery, Brain Tumor Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Doo-Sik Kong
- Department of Neurosurgery, Brain Tumor Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Jung Won Choi
- Department of Neurosurgery, Brain Tumor Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Kyuha Chong
- Department of Neurosurgery, Brain Tumor Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Won Jae Lee
- Department of Neurosurgery, Brain Tumor Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Severance Hospital, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Tumor Center, Severance Hospital, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seok-Gu Kang
- Department of Neurosurgery, Severance Hospital, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Tumor Center, Severance Hospital, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ju Hyung Moon
- Department of Neurosurgery, Severance Hospital, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Tumor Center, Severance Hospital, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jaeho Cho
- Department of Radiation Oncology, Heavy Ion Therapy Research Institute, Yonsei Cancer Center, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Hong In Yoon
- Department of Radiation Oncology, Heavy Ion Therapy Research Institute, Yonsei Cancer Center, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
23
|
Niu Z, Yang Z, Sun S, Zeng Z, Han Q, Wu L, Bai J, Li H, Xia H. Clinical analysis of the efficacy of radiation therapy for primary high-grade gliomas guided by biological rhythms. Transl Oncol 2024; 45:101973. [PMID: 38705052 PMCID: PMC11089398 DOI: 10.1016/j.tranon.2024.101973] [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: 12/29/2023] [Revised: 04/05/2024] [Accepted: 04/20/2024] [Indexed: 05/07/2024] Open
Abstract
OBJECTIVE High-grade glioma (HGG) patients frequently encounter treatment resistance and relapse, despite numerous interventions seeking enhanced survival outcomes yielding limited success. Consequently, this study, rooted in our prior research, aimed to ascertain whether leveraging circadian rhythm phase attributes could optimize radiotherapy results. METHODS In this retrospective analysis, we meticulously selected 121 HGG cases with synchronized rhythms through Cosinor analysis. Post-surgery, all subjects underwent standard radiotherapy alongside Temozolomide chemotherapy. Random allocation ensued, dividing patients into morning (N = 69) and afternoon (N = 52) radiotherapy cohorts, enabling a comparison of survival and toxicity disparities. RESULTS The afternoon radiotherapy group exhibited improved overall survival (OS) and progression-free survival (PFS) relative to the morning cohort. Notably, median OS extended to 25.6 months versus 18.5 months, with P = 0.014, with median PFS at 20.6 months versus 13.3 months, with P = 0.022, post-standardized radiotherapy. Additionally, lymphocyte expression levels in the afternoon radiation group 32.90(26.10, 39.10) significantly exceeded those in the morning group 31.30(26.50, 39.20), with P = 0.032. CONCLUSIONS This study underscores the markedly prolonged average survival within the afternoon radiotherapy group. Moreover, lymphocyte proportion demonstrated a notable elevation in the afternoon group. Timely and strategic adjustments of therapeutic interventions show the potential to improve therapeutic efficacy, while maintaining vigilant systemic immune surveillance. A comprehensive grasp of physiological rhythms governing both the human body and tumor microenvironment can refine treatment efficacy, concurrently curtailing immune-related damage-a crucial facet of precision medicine.
Collapse
Affiliation(s)
- Zhanfeng Niu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Zhihua Yang
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Shengyu Sun
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Zhong Zeng
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China; Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, PR China
| | - Qian Han
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China; Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, PR China
| | - Liang Wu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Jinbo Bai
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Hailiang Li
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Hechun Xia
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China; Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, PR China.
| |
Collapse
|
24
|
Kim S, Byun HK, Shin J, Lee IJ, Sung W. Normal Tissue Complication Probability Modeling of Severe Radiation-Induced Lymphopenia Using Blood Dose for Patients With Hepatocellular Carcinoma. Int J Radiat Oncol Biol Phys 2024; 119:1011-1020. [PMID: 38056776 DOI: 10.1016/j.ijrobp.2023.11.060] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/24/2023] [Accepted: 11/25/2023] [Indexed: 12/08/2023]
Abstract
PURPOSE This study aimed to develop a normal tissue complication probability (NTCP) model to estimate the risk of severe radiation-induced lymphopenia (SRIL; absolute lymphocyte count [ALC] < 500/μL) by using the blood dose of patients with hepatocellular carcinoma (HCC). METHODS AND MATERIALS We retrospectively collected data from 75 patients with HCC who received radiation therapy (RT) between 2015 and 2018. The hematological dose framework calculated blood dose-volume histograms (DVHs) using a predefined blood flow model, organ DVHs, the number of treatment fractions, and beam delivery time. A Lyman-Kutcher-Burman model with a generalized equivalent dose was used to establish the NTCP model, reflecting the whole-blood DVHs. Optimization of the Lyman-Kutcher-Burman parameters was conducted by minimizing a negative log-likelihood function. RESULTS There were 6, 4, 18, 33, and 14 patients in the groups with radiation-induced lymphopenia grades 0, 1, 2, 3, and 4, respectively. The median pre- and post-RT ALC values were 1410/μL (range, 520-3710/μL) and 470/μL (range, 60-1760/μL), respectively. There was a correlation between mean blood dose and ALC depletion (Pearson r = -0.664; P < .001). The average mean blood doses in each radiation-induced lymphopenia group were 2.90 Gy (95% CI, 1.96-3.85 Gy) for grade 0 to 1, 5.29 Gy (95% CI, 4.12-6.45 Gy) for grade 2, 8.81 Gy (95% CI, 7.55-10.07 Gy) for grade 3, and 11.69 Gy (95% CI, 9.82-17.57 Gy) for grade 4. When applying the developed NTCP model to predict SRIL, the area under the receiver operating characteristic curve and Brier score values were 0.89 and 0.12, respectively. CONCLUSIONS We developed the first NTCP model based on whole-blood DVHs for estimating SRIL after abdominal RT in patients with HCC. Our results showed a strong correlation between blood dose and ALC depletion, suggesting the potential to predict the risk of SRIL occurrence using blood dose.
Collapse
Affiliation(s)
- Seohan Kim
- Deparments of Biomedical Engineering and Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hwa Kyung Byun
- Department of Radiation Oncology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, South Korea
| | - Jungwook Shin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Ik Jae Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea.
| | - Wonmo Sung
- Deparments of Biomedical Engineering and Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
| |
Collapse
|
25
|
Tubin S. A Partial Tumor Irradiation Approach for Complex Bulky Disease. Semin Radiat Oncol 2024; 34:323-336. [PMID: 38880541 DOI: 10.1016/j.semradonc.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
A large proportion of cancer patients present with unresectable bulky disease at baseline or following treatment failure. The data available in the literature suggest that the vast majority of these patients do not benefit from available standard therapies. Therefore the clinical outcomes are poor; patients are desperate and usually relegated to palliative or best supportive care as the only options. Large tumor masses are usually hypoxic, resistant to radiation and systemic therapy, with extensive regional infiltration of the surrounding critical organs, the presence of which makes it impossible to deliver a radical dose of radiation. Promising data in terms of improved therapeutic ratio where such complex tumors are concerned can be seen with the use of new emerging unconventional radiotherapy techniques known as spatially fractionated radiotherapies (SFRT). One of them is PATHY, or PArtial Tumor irradiation targeting HYpoxic segment, which is characterized by a very short treatment course offering a large spectrum of therapeutic benefits in terms of the symptom relief, quality of life, local tumor control, neoadjuvant and immunomodulatory effects.
Collapse
Affiliation(s)
- Slavisa Tubin
- Medaustron Center for Ion Therapy, Marie-Curie Strasse 5, Wiener Neustadt 2700, Austria; Heidelberg University Hospital, Department of Radiation Oncology and Radiation Therapy, Im Neuenheimer Feld 400 69120 Heidelberg; Montefiore Medical Center Radiation Oncology, 111 E 210th St, New York, NY, United States.
| |
Collapse
|
26
|
Bergerud KMB, Berkseth M, Pardoll DM, Ganguly S, Kleinberg LR, Lawrence J, Odde DJ, Largaespada DA, Terezakis SA, Sloan L. Radiation Therapy and Myeloid-Derived Suppressor Cells: Breaking Down Their Cancerous Partnership. Int J Radiat Oncol Biol Phys 2024; 119:42-55. [PMID: 38042450 PMCID: PMC11082936 DOI: 10.1016/j.ijrobp.2023.11.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
Radiation therapy (RT) has been a primary treatment modality in cancer for decades. Increasing evidence suggests that RT can induce an immunosuppressive shift via upregulation of cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs). MDSCs inhibit antitumor immunity through potent immunosuppressive mechanisms and have the potential to be crucial tools for cancer prognosis and treatment. MDSCs interact with many different pathways, desensitizing tumor tissue and interacting with tumor cells to promote therapeutic resistance. Vascular damage induced by RT triggers an inflammatory signaling cascade and potentiates hypoxia in the tumor microenvironment (TME). RT can also drastically modify cytokine and chemokine signaling in the TME to promote the accumulation of MDSCs. RT activation of the cGAS-STING cytosolic DNA sensing pathway recruits MDSCs through a CCR2-mediated mechanism, inhibiting the production of type 1 interferons and hampering antitumor activity and immune surveillance in the TME. The upregulation of hypoxia-inducible factor-1 and vascular endothelial growth factor mobilizes MDSCs to the TME. After recruitment, MDSCs promote immunosuppression by releasing reactive oxygen species and upregulating nitric oxide production through inducible nitric oxide synthase expression to inhibit cytotoxic activity. Overexpression of arginase-1 on subsets of MDSCs degrades L-arginine and downregulates CD3ζ, inhibiting T-cell receptor reactivity. This review explains how radiation promotes tumor resistance through activation of immunosuppressive MDSCs in the TME and discusses current research targeting MDSCs, which could serve as a promising clinical treatment strategy in the future.
Collapse
Affiliation(s)
| | - Matthew Berkseth
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sudipto Ganguly
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lawrence R Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, Minnesota
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - David A Largaespada
- Departments of Pediatrics and Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota
| | | | - Lindsey Sloan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
27
|
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
|
28
|
Wisdom AJ, Barker CA, Chang JY, Demaria S, Formenti S, Grassberger C, Gregucci F, Hoppe BS, Kirsch DG, Marciscano AE, Mayadev J, Mouw KW, Palta M, Wu CC, Jabbour SK, Schoenfeld JD. The Next Chapter in Immunotherapy and Radiation Combination Therapy: Cancer-Specific Perspectives. Int J Radiat Oncol Biol Phys 2024; 118:1404-1421. [PMID: 38184173 DOI: 10.1016/j.ijrobp.2023.12.046] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/20/2023] [Accepted: 12/30/2023] [Indexed: 01/08/2024]
Abstract
Immunotherapeutic agents have revolutionized cancer treatment over the past decade. However, most patients fail to respond to immunotherapy alone. A growing body of preclinical studies highlights the potential for synergy between radiation therapy and immunotherapy, but the outcomes of clinical studies have been mixed. This review summarizes the current state of immunotherapy and radiation combination therapy across cancers, highlighting existing challenges and promising areas for future investigation.
Collapse
Affiliation(s)
- Amy J Wisdom
- Harvard Radiation Oncology Program, Boston, Massachusetts
| | - Christopher A Barker
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joe Y Chang
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Clemens Grassberger
- Department of Radiation Oncology, University of Washington, Fred Hutch Cancer Center, Seattle, Washington
| | - Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Bradford S Hoppe
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida
| | - David G Kirsch
- Department of Radiation Oncology, University of Toronto, Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ariel E Marciscano
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jyoti Mayadev
- Department of Radiation Oncology, UC San Diego School of Medicine, San Diego, California
| | - Kent W Mouw
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Manisha Palta
- Department of Radiation Oncology, Duke Cancer Center, Durham, North Carolina
| | - Cheng-Chia Wu
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
| | - Jonathan D Schoenfeld
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
29
|
Saeed AM, Bentzen SM, Ahmad H, Pham L, Woodworth GF, Mishra MV. Systematic review and pooled analysis of the impact of treatment-induced lymphopenia on survival of glioblastoma patients. Radiat Oncol 2024; 19:36. [PMID: 38481255 PMCID: PMC10938829 DOI: 10.1186/s13014-023-02393-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/17/2023] [Indexed: 03/17/2024] Open
Abstract
PURPOSE/OBJECTIVE(S) Treatment related lymphopenia is a known toxicity for glioblastoma (GBM) patients and several single-institution studies have linked lymphopenia with poor survival outcomes. We performed a systematic review and pooled analysis to evaluate the association between lymphopenia and overall survival (OS) for GBM patients undergoing chemotherapy and radiation therapy (RT). MATERIALS/METHODS Following PRISMA guidelines, a systematic literature review of the MEDLINE database and abstracts from ASTRO, ASCO, and SNO annual meetings was conducted. A pooled analysis was performed using inverse variance-weighted random effects to generate a pooled estimate of the hazard ratio of association between lymphopenia and OS. RESULTS Ten of 104 identified studies met inclusion criteria, representing 1,718 patients. The lymphopenia cutoff value varied (400-1100 cells/uL) and as well as the timing of its onset. Studies were grouped as time-point (i.e., lymphopenia at approximately 2-months post-RT) or time-range (any lymphopenia occurrence from treatment-start to approximately 2-months post-RT. The mean overall pooled incidence of lymphopenia for all studies was 31.8%, and 11.8% vs. 39.9% for time-point vs. time-range studies, respectively. Lymphopenia was associated with increased risk of death, with a pooled HR of 1.78 (95% CI 1.46-2.17, P < 0.00001) for the time-point studies, and a pooled HR of 1.38 (95% CI 1.24-1.55, P < 0.00001) for the time-point studies. There was no significant heterogeneity between studies. CONCLUSION These results strengthen observations from previous individual single-institution studies and better defines the magnitude of the association between lymphopenia with OS in GBM patients, highlighting lymphopenia as a poor prognostic factor.
Collapse
Affiliation(s)
- Ali M Saeed
- Department of Radiation Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA
- Maryland Proton Treatment Center, Baltimore, MD, USA
| | - Søren M Bentzen
- Department of Radiation Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA
- Department of Epidemiology and Public Health, Division of Biostatistics and Bioinformatics, University of Maryland School of Medicine, Baltimore, USA
| | - Haroon Ahmad
- Department of Medical Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA
| | - Lily Pham
- Department of Medical Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mark V Mishra
- Department of Radiation Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA.
- Maryland Proton Treatment Center, Baltimore, MD, USA.
| |
Collapse
|
30
|
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
|
31
|
Thompson EM, Kang KD, Stevenson K, Zhang H, Gromeier M, Ashley D, Brown M, Friedman GK. Elucidating cellular response to treatment with viral immunotherapies in pediatric high-grade glioma and medulloblastoma. Transl Oncol 2024; 40:101875. [PMID: 38183802 PMCID: PMC10809117 DOI: 10.1016/j.tranon.2024.101875] [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: 07/19/2023] [Revised: 12/08/2023] [Accepted: 12/27/2023] [Indexed: 01/08/2024] Open
Abstract
HSV G207, a double-stranded, DNA virus, and the polio:rhinovirus chimera, PVSRIPO, a single positive-strand RNA virus, are viral immunotherapies being used to treat pediatric malignant brain tumors in clinical trials. The purpose of this work is to elucidate general response patterns and putative biomarkers of response. Multiple pediatric high-grade glioma and medulloblastoma cell lines were treated with various multiplicities of infection of G207 or PVSRIPO. There was a significant inverse correlation between expression of one HSV cellular receptor, CD111, and the lethal dose of 50% of cells (LD50) of cells treated with G207 (r = -0.985, P<0.001) but no correlation between PVSRIPO cellular receptor expression (CD155) and LD50. RNA sequencing of control cells and cells treated for 8 and 24 h revealed that there were few shared differentially expressed (DE) genes between cells treated with PVSRIPO and G207: GCLM, LANCL2, and RBM3 were enriched whilst ADAMTS1 and VEGFA were depleted. Likewise, there were few shared DE genes enriched between medulloblastoma and high-grade glioma cell lines treated with G207: GPSM2, CHECK2, SEPTIN2, EIF4G2, GCLM, GDAP1, LANCL2, and PWP1. Treatment with G207 and PVSRIPO appear to cause disparate gene enrichment and depletion suggesting disparate molecular mechanisms in malignant pediatric brain tumors.
Collapse
Affiliation(s)
- Eric M Thompson
- Department of Neurosurgery, University of Chicago, Chicago, IL, USA; Department of Neurosurgery, Duke University, Durham, NC, USA.
| | - Kyung-Don Kang
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA; The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kevin Stevenson
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Hengshan Zhang
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | | | - David Ashley
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Michael Brown
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Gregory K Friedman
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA; The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
32
|
Park JH, Lee SW, Choi D, Lee C, Sung YC. Harnessing the Power of IL-7 to Boost T Cell Immunity in Experimental and Clinical Immunotherapies. Immune Netw 2024; 24:e9. [PMID: 38455462 PMCID: PMC10917577 DOI: 10.4110/in.2024.24.e9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/09/2024] Open
Abstract
The cytokine IL-7 plays critical and nonredundant roles in T cell immunity so that the abundance and availability of IL-7 act as key regulatory mechanisms in T cell immunity. Importantly, IL-7 is not produced by T cells themselves but primarily by non-lymphoid lineage stromal cells and epithelial cells that are limited in their numbers. Thus, T cells depend on cell extrinsic IL-7, and the amount of in vivo IL-7 is considered a major factor in maximizing and maintaining the number of T cells in peripheral tissues. Moreover, IL-7 provides metabolic cues and promotes the survival of both naïve and memory T cells. Thus, IL-7 is also essential for the functional fitness of T cells. In this regard, there has been an extensive effort trying to increase the protein abundance of IL-7 in vivo, with the aim to augment T cell immunity and harness T cell functions in anti-tumor responses. Such approaches started under experimental animal models, but they recently culminated into clinical studies, with striking effects in re-establishing T cell immunity in immunocompromised patients, as well as boosting anti-tumor effects. Depending on the design, glycosylation, and the structure of recombinantly engineered IL-7 proteins and their mimetics, recombinant IL-7 molecules have shown dramatic differences in their stability, efficacy, cellular effects, and overall immune functions. The current review is aimed to summarize the past and present efforts in the field that led to clinical trials, and to highlight the therapeutical significance of IL-7 biology as a master regulator of T cell immunity.
Collapse
Affiliation(s)
- Jung-Hyun Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Donghoon Choi
- Research Institute of NeoImmune Tech., Co, Ltd., Bio Open Innovation Center, Pohang 37666, Korea
| | - Changhyung Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Young Chul Sung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| |
Collapse
|
33
|
Wang H, Li Y, Hu P, Zhang J. The Correlation Between Low-Dose Radiotherapy Area of the Mediastinum and CD8+T Cells and the Efficacy of Radiotherapy for Non-Small Cell Lung Cancer. Cancer Manag Res 2024; 16:23-35. [PMID: 38230351 PMCID: PMC10790660 DOI: 10.2147/cmar.s438440] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 01/08/2024] [Indexed: 01/18/2024] Open
Abstract
Background Radiation therapy (RT) can cause changes in peripheral blood immune cells. The relationship between the efficacy of radiation therapy for non-small cell lung cancer (NSCLC) and immune cell changes and the study of how mediastinal radiation dose parameters affect immune cell changes is still unclear. This study aims to analyze the relationship between immune cell changes induced by radiotherapy and the efficacy of NSCLC radiotherapy, as well as the relationship between radiotherapy dose parameters and immune cell changes. Materials and Methods We retrospectively analyzed the data of NSCLC patients receiving mediastinal radiation therapy from 2020 to 2022. Collect lymphocytes and circulating immune cells within one week before and after radiotherapy and collect the dose-volume parameters of the whole mediastinum in the patient's RT planning system. Analyze the changes in lymphocytes and radiotherapy effects after radiotherapy, and explore the relationship between radiotherapy dose parameters and immune cell changes. Results A total of 72 patients were enrolled. Compared with before radiotherapy, the proportion of CD3+T cells, CD8+T cells, and CD8/Treg in peripheral blood significantly increased after radiotherapy (P<0.05). The increase in CD8+T cells and CD8/Treg after radiotherapy was correlated with Objective response rate (ORR) (P<0.05). Based on binary logistic univariate and multivariate regression analysis, an increase in CD8+T cells after radiotherapy is an independent predictor of objective tumor response after radiotherapy (OR=12.71, 95% CI=3.64-44.64, P=0.01), and Volume of 200 cGy irradiation (V2) is an independent positive predictor of an increase in CD8+T lymphocyte ratio after radiotherapy (high group, OR=3.40, 95% CI=1.13-10.36, P=0.03). Conclusion The increase in CD8+T cells after radiotherapy can positively predict the short-term efficacy of radiotherapy. Mediastinal low-dose radiation therapy can increase CD8+T cells, thereby improving the short-term efficacy of radiotherapy. These potentially related mechanisms are worth further verification and exploration.
Collapse
Affiliation(s)
- Hang Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Lung Cancer Institute, Jinan, Shandong Province, 25000, People’s Republic of China
| | - Yang Li
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Lung Cancer Institute, Jinan, Shandong Province, 25000, People’s Republic of China
| | - Pingping Hu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Lung Cancer Institute, Jinan, Shandong Province, 25000, People’s Republic of China
| | - Jiandong Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Lung Cancer Institute, Jinan, Shandong Province, 25000, People’s Republic of China
| |
Collapse
|
34
|
Qu F, Luo Y, Peng Y, Yu H, Sun L, Liu S, Zeng X. Construction and validation of a prognostic nutritional index-based nomogram for predicting pathological complete response in breast cancer: a two-center study of 1,170 patients. Front Immunol 2024; 14:1335546. [PMID: 38274836 PMCID: PMC10808698 DOI: 10.3389/fimmu.2023.1335546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
Background Pathological complete response (pCR) after neoadjuvant chemotherapy (NAC) is associated with favorable outcomes in breast cancer patients. Identifying reliable predictors for pCR can assist in selecting patients who will derive the most benefit from NAC. The prognostic nutritional index (PNI) serves as an indicator of nutritional status and systemic immune competence. It has emerged as a prognostic biomarker in several malignancies; however, its predictive value for pCR in breast cancer remains uncertain. The objective of this study is to assess the predictive value of pretreatment PNI for pCR in breast cancer patients. Methods A total of 1170 patients who received NAC in two centers were retrospectively analyzed. The patients were divided into three cohorts: a training cohort (n=545), an internal validation cohort (n=233), and an external validation cohort (n=392). Univariate and multivariate analyses were performed to assess the predictive value of PNI and other clinicopathological factors. A stepwise logistic regression model for pCR based on the smallest Akaike information criterion was utilized to develop a nomogram. The C-index, calibration plots and decision curve analysis (DCA) were used to evaluate the discrimination, calibration and clinical value of the model. Results Patients with a high PNI (≥53) had a significantly increased pCR rate (OR 2.217, 95% CI 1.215-4.043, p=0.009). Tumor size, clinical nodal status, histological grade, ER, Ki67 and PNI were identified as independent predictors and included in the final model. A nomogram was developed as a graphical representation of the model, which incorporated the PNI and five other factors (AIC=356.13). The nomogram demonstrated satisfactory calibration and discrimination in the training cohort (C-index: 0.816, 95% CI 0.765-0.866), the internal validation cohort (C-index: 0.780, 95% CI 0.697-0.864) and external validation cohort (C-index: 0.714, 95% CI 0.660-0.769). Furthermore, DCA indicated a clinical net benefit from the nomogram. Conclusion The pretreatment PNI is a reliable predictor for pCR in breast cancer patients. The PNI-based nomogram is a low-cost, noninvasive tool with favorable predictive accuracy for pCR, which can assist in determining individualized treatment strategies for breast cancer patients.
Collapse
Affiliation(s)
- Fanli Qu
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaxi Luo
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Peng
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haochen Yu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lu Sun
- Department of Thyroid and Breast Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Shengchun Liu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaohua Zeng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
35
|
Kim YH, Choi YD, Ahn SJ, Kim YC, Oh IJ, Nam TK, Jeong JU, Song JY. Blood Lymphocytes as a Prognostic Factor for Stage III Non-Small Cell Lung Cancer with Concurrent Chemoradiation. Chonnam Med J 2024; 60:40-50. [PMID: 38304134 PMCID: PMC10828085 DOI: 10.4068/cmj.2024.60.1.40] [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: 10/25/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 02/03/2024] Open
Abstract
We aimed to identify blood lymphocytes as a prognostic factor for survival in patients with locally advanced stage III non-small cell lung cancer (NSCLC) treated with concurrent chemoradiotherapy (CCRT). This is a secondary study of 196 patients enrolled in the Korean Radiation Oncology Group 0903 phase III clinical trial to evaluate the prognostic significance of circulating blood lymphocyte levels. The median total lymphocyte count (TLC) reduction ratio during CCRT was 0.74 (range: 0.29-0.97). In multivariate analysis, patient age (p=0.014) and gross tumor volume (GTV, p=0.031) were significant factors associated with overall survival, while TLC reduction (p=0.018) and pretreatment neutrophil-to-lymphocyte ratio (NLR; p=0.010) were associated with progression-free survival (PFS). In multivariate logistic regression analysis, pretreatment NLR, GTV, and heart V20 were significantly associated with TLC reduction. Immunohistochemical analysis of programmed death ligand 1 and CD8 expression on T cells was performed on 84 patients. CD8 expression was not significantly associated with the pretreatment lymphocyte count (p=0.673), and PDL1 expression was not significantly associated with OS or PFS. Univariate analysis revealed that high CD8 expression in TILs was associated with favorable OS and was significantly associated with favorable PFS (p=0.032). TLC reduction during CCRT is a significant prognostic factor for PFS, and heart V20 is significantly associated with TLC reduction. Thus, in the era of immunotherapy, constraining the volume of the radiation dose to the whole heart must be prioritized for the better survival outcomes.
Collapse
Affiliation(s)
- Yong-Hyub Kim
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Yoo-Duk Choi
- Department of Pathology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Sung-Ja Ahn
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Young-Chul Kim
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - In-Jae Oh
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Taek-Keun Nam
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Jae-Uk Jeong
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Ju-Young Song
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
36
|
Jiang J, Zhai R, Kong F, Du C, Ying H. Nomograms containing body dose parameters for predicting survival in patients with nasopharyngeal carcinoma. Eur Arch Otorhinolaryngol 2024; 281:181-192. [PMID: 37552282 PMCID: PMC10764493 DOI: 10.1007/s00405-023-08173-9] [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: 06/09/2023] [Accepted: 07/31/2023] [Indexed: 08/09/2023]
Abstract
PURPOSE To assess the impact of body dose on survival outcomes in nasopharyngeal carcinoma (NPC) patients and to create novel nomograms incorporating body dose parameters for predicting survival. METHODS 594 of non-metastasis NPC patients (training group, 396; validation group, 198) received intensity-modulated radiation therapy at our institution from January 2012 to December 2016. Patient characteristics, body dose parameters in dose-volume histogram (DVH) and hematology profiles were collected for predicting overall survival (OS) and progression-free survival (PFS). Nomograms for OS and PFS were developed using the selected predictors. Each nomogram was evaluated based on its C-index and calibration curve. RESULTS Body dose-based risk score for OS (RSOS), N stage, age, and induction chemotherapy were independent predictors for OS, with a C-index of 0.784 (95% CI 0.749-0.819) in the training group and 0.763 (95% CI 0.715-0.810) in the validation group for the nomogram. As for PFS, the most important predictors were the body dose-based risk score for PFS (RSPFS), N stage, and induction chemotherapy. C-index of PFS nomogram was 0.706 (95% CI 0.681-0.720) in the training group and 0.691 (95% CI 0.662-0.711) in the validation group. The two models outperformed the TNM staging system in predicting outcomes. CONCLUSIONS Body dose coverage is a useful predictor of prognosis in clinical routine patients. The novel nomograms integrating body dose parameters can precisely predict OS and PFS in NPC patients.
Collapse
Affiliation(s)
- Jianyun Jiang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Ruiping Zhai
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Fangfang Kong
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Chengrun Du
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Hongmei Ying
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China.
| |
Collapse
|
37
|
Valdes Angues R, Perea Bustos Y. SARS-CoV-2 Vaccination and the Multi-Hit Hypothesis of Oncogenesis. Cureus 2023; 15:e50703. [PMID: 38234925 PMCID: PMC10792266 DOI: 10.7759/cureus.50703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 01/19/2024] Open
Abstract
Cancer is a complex and dynamic disease. The "hallmarks of cancer" were proposed by Hanahan and Weinberg (2000) as a group of biological competencies that human cells attain as they progress from normalcy to neoplastic transformation. These competencies include self-sufficiency in proliferative signaling, insensitivity to growth-suppressive signals and immune surveillance, the ability to evade cell death, enabling replicative immortality, reprogramming energy metabolism, inducing angiogenesis, and activating tissue invasion and metastasis. Underlying these competencies are genome instability, which expedites their acquisition, and inflammation, which fosters their function(s). Additionally, cancer exhibits another dimension of complexity: a heterogeneous repertoire of infiltrating and resident host cells, secreted factors, and extracellular matrix, known as the tumor microenvironment, that through a dynamic and reciprocal relationship with cancer cells supports immortality, local invasion, and metastatic dissemination. This staggering intricacy calls for caution when advising all people with cancer (or a previous history of cancer) to receive the COVID-19 primary vaccine series plus additional booster doses. Moreover, because these patients were not included in the pivotal clinical trials, considerable uncertainty remains regarding vaccine efficacy, safety, and the risk of interactions with anticancer therapies, which could reduce the value and innocuity of either medical treatment. After reviewing the available literature, we are particularly concerned that certain COVID-19 vaccines may generate a pro-tumorigenic milieu (i.e., a specific environment that could lead to neoplastic transformation) that predisposes some (stable) oncologic patients and survivors to cancer progression, recurrence, and/or metastasis. This hypothesis is based on biological plausibility and fulfillment of the multi-hit hypothesis of oncogenesis (i.e., induction of lymphopenia and inflammation, downregulation of angiotensin-converting enzyme 2 (ACE2) expression, activation of oncogenic cascades, sequestration of tumor suppressor proteins, dysregulation of the RNA-G quadruplex-protein binding system, alteration of type I interferon responses, unsilencing of retrotransposable elements, etc.) together with growing evidence and safety reports filed to Vaccine Adverse Effects Report System (VAERS) suggesting that some cancer patients experienced disease exacerbation or recurrence following COVID-19 vaccination. In light of the above and because some of these concerns (i.e., alteration of oncogenic pathways, promotion of inflammatory cascades, and dysregulation of the renin-angiotensin system) also apply to cancer patients infected with SARS-CoV-2, we encourage the scientific and medical community to urgently evaluate the impact of both COVID-19 and COVID-19 vaccination on cancer biology and tumor registries, adjusting public health recommendations accordingly.
Collapse
Affiliation(s)
- Raquel Valdes Angues
- Neurology, Oregon Health and Science University School of Medicine, Portland, USA
| | | |
Collapse
|
38
|
Yang G, Yoon HI, Lee J, Kim J, Kim H, Cho J, Lee CG, Chang JS, Cho Y, Kim JS, Kim KH. Risk of on-treatment lymphopenia is associated with treatment outcome and efficacy of consolidation immunotherapy in patients with non-small cell lung cancer treated with concurrent chemoradiotherapy. Radiother Oncol 2023; 189:109934. [PMID: 37783291 DOI: 10.1016/j.radonc.2023.109934] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/04/2023]
Abstract
BACKGROUND AND PURPOSE The ability of the effective dose to immune cells (EDIC) and the pre-radiotherapy (RT) absolute lymphocyte count (ALC) to predict lymphopenia during RT, treatment outcomes, and efficacy of consolidation immunotherapy in patients with locally advanced non-small cell lung cancer was investigated. METHODS AND MATERIALS Among 517 patients treated with concurrent chemoradiotherapy, EDIC was calculated using the mean doses to the lungs, heart, and total body. The patients were grouped according to high and low EDIC and pre-RT ALC, and the correlations with radiation-induced lymphopenia and survival outcomes were determined. RESULTS Altogether, 195 patients (37.7%) received consolidation immunotherapy. The cutoff values of EDIC and pre-RT ALC for predicting severe lymphopenia were 2.89 Gy and 2.03 × 109 cells/L, respectively. The high-risk group was defined as EDIC ≥ 2.89 Gy and pre-RT ALC < 2.03 × 109 cells/L, while the low-risk group as EDIC < 2.89 Gy and pre-RT ALC ≥ 2.03 × 109 cells/L, and the rest of the patients as the intermediate-risk group. The incidences of severe lymphopenia during RT in the high-, intermediate-, and low-risk groups were 90.1%, 77.1%, and 52.3%, respectively (P < 0.001). The risk groups could independently predict both progression-free (P < 0.001) and overall survival (P < 0.001). The high-risk group showed a higher incidence of locoregional and distant recurrence (P < 0.001). Consolidation immunotherapy showed significant survival benefit in the low- and intermediate-risk groups but not in the high-risk group. CONCLUSIONS The combination of EDIC and pre-RT ALC predicted severe lymphopenia, recurrence, and survival. It may potentially serve as a biomarker for consolidation immunotherapy.
Collapse
Affiliation(s)
- Gowoon Yang
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hong In Yoon
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Joongyo Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jihun Kim
- Department of Radiation Oncology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eon-ju-ro, Gangnam-gu, Seoul 06273, Republic of Korea
| | - Hojin Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jaeho Cho
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Chang Geol Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jee Suk Chang
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Yeona Cho
- Department of Radiation Oncology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eon-ju-ro, Gangnam-gu, Seoul 06273, Republic of Korea
| | - Jin Sung Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Kyung Hwan Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
39
|
Strati P, Spiotto MT. Incorporating Immunotherapy with Radiotherapy for Lymphomas. LYMPHATICS 2023; 1:273-286. [PMID: 39917366 PMCID: PMC11800356 DOI: 10.3390/lymphatics1030018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Radiotherapy and/or chemotherapy have been used for nearly 100 years to treat lymphoma. Recently, immunotherapy has been incorporated into the treatment of lymphomas. Here, we will review both the role of immunotherapy in lymphoma as well as the feasibility of incorporating immunotherapies with conventional lymphoma treatments, especially radiotherapy. Immunotherapy agents include checkpoint inhibitors that target the PD-1/PD-L1 axis, CTLA-4, or CD47. In addition, other immunotherapy agents such as bi-specific antibodies and CD19 CAR-T cell therapy are being implemented in various non-Hodgkin's lymphomas. Extrapolating from observations in other disease sites and incorporating immunotherapy with conventional treatments of lymphoma, including radiotherapy, may have opposing effects. Radiotherapy may stimulate anti-tumor immune responses that synergize with immunotherapies. In contrast, radiotherapy, as well as chemotherapy, may also induce local and systemic immune dysfunction which reduces the efficacy of immunotherapies. With newer radiation treatment techniques and limited radiation fields, it is likely that the efficacy of immunotherapy can be maintained when included with conventional treatments. Therefore, there remains an unmet need to better understand the role of immunotherapy alone and in combination with current treatments in lymphoma patients.
Collapse
Affiliation(s)
- Paolo Strati
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael T. Spiotto
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
40
|
Tefferi A, Loscocco GG, Farrukh F, Szuber N, Mannelli F, Pardanani A, Hanson CA, Ketterling RP, De Stefano V, Carobbio A, Barbui T, Guglielmelli P, Gangat N, Vannucchi AM. A globally applicable "triple A" risk model for essential thrombocythemia based on Age, Absolute neutrophil count, and Absolute lymphocyte count. Am J Hematol 2023; 98:1829-1837. [PMID: 37665758 DOI: 10.1002/ajh.27079] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023]
Abstract
We examined the individual prognostic contribution of absolute neutrophil (ANC), lymphocyte (ALC), and monocyte (AMC) counts, on overall (OS), leukemia-free (LFS), and myelofibrosis-free (MFFS) survival in essential thrombocythemia (ET). Informative cases (N = 598; median age 59 years; females 62%) were retrospectively accrued from a Mayo Clinic database: JAK2 59%, CALR 27%, triple-negative 11%, and MPL 3%; international prognostic scoring system for ET (IPSET) risk high 21%, intermediate 42%, and low 37%; 7% (37/515) had abnormal karyotype and 10% (21/205) adverse mutations (SF3B1/SRSF2/U2AF1/TP53). At median 8.4 years, 163 (27%) deaths, 71 (12%) fibrotic, and 20 (3%) leukemic transformations were recorded. Multivariable analysis resulted in HR (95% CI) of 16.5 (9.9-27.4) for age > 70 years, 3.7 (2.3-6.0) for age 50-70 years, 2.4 (1.7-3.3) for ANC ≥8 × 109 /L, and 1.9 (1.4-2.6) for ALC <1.7 × 109 /L. The corresponding HR-based scores were 4, 2, 1, and 1, resulting in an new 4-tiered AgeAncAlc (AAA; triple A) risk model: high (5-6 points; median survival 8 years; HR 30.1, 95% CI 17.6-54), intermediate-2 (4 points; median 13.5 years; HR 12.7, 95% CI 7.1-23.0), intermediate-1 (2-3 points; median 20.7 years; HR 3.8, 95% CI 2.3-6.4) and low (0-1 points; median 47 years). The AAA model (Akaike Information Criterion [AIC] 621) performed better than IPSET (AIC 647) and was subsequently validated by an external University of Florence ET cohort (N = 485). None of the AAA variables predicted LFS while ALC <1.7 × 109 /L was associated with inferior MFFS (p = .01). Adverse mutations (p < .01) and karyotype (p < .01) displayed additional prognostic value without disqualifying the prognostic integrity of the AAA model. This study proposes a simple and globally applicable survival model for ET, which can be used as a platform for further molecular refinement. This study also suggests a potential role for immune-related biomarkers, as a prognostic tool in myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Divisions of Hematology and Hematopathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Giuseppe G Loscocco
- Department of Experimental and Clinical Medicine, CRIMM, Center Research, and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| | - Faiqa Farrukh
- Divisions of Hematology and Hematopathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Natasha Szuber
- Department of Hematology, Université de Montréal, Quebec, Canada
| | - Francesco Mannelli
- Department of Experimental and Clinical Medicine, CRIMM, Center Research, and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| | - Animesh Pardanani
- Divisions of Hematology and Hematopathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Curtis A Hanson
- Divisions of Hematology and Hematopathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Rhett P Ketterling
- Divisions of Hematology and Hematopathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Valerio De Stefano
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Rome, Italy
| | | | - Tiziano Barbui
- FROM esearch Foundation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Paola Guglielmelli
- Department of Experimental and Clinical Medicine, CRIMM, Center Research, and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| | - Naseema Gangat
- Divisions of Hematology and Hematopathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Alessandro M Vannucchi
- Department of Experimental and Clinical Medicine, CRIMM, Center Research, and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| |
Collapse
|
41
|
Jackman RP, Darst O, Gaillard B, Tran JQ, Tomayko MM, Muench MO. Enhanced alloresponse to platelet transfusion due to immune dysregulation following ablative chemotherapy in mice. Front Immunol 2023; 14:1281123. [PMID: 38090570 PMCID: PMC10711281 DOI: 10.3389/fimmu.2023.1281123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Alloimmunization is common following platelet transfusion and can result in negative outcomes for recipients such as refractoriness to subsequent transfusions and rejection of transplants. Healthy people do not receive blood transfusions, and the diseases and therapies that result in a need to transfuse have significant impacts on the immunological environment to which these alloantigens are introduced. Ablative chemotherapies are common among platelet recipients and have potent immunological effects. In this study, we modeled the impact of chemotherapy on the alloresponse to platelet transfusion. As chemotherapies are generally regarded as immunosuppressive, we hypothesized that that they would result in a diminished alloresponse. Methods Mice were given a combination chemotherapeutic treatment of cytarabine and doxorubicin followed by transfusion of allogeneic platelets, and compared to controls given no treatment, chemotherapy alone, or transfusion alone. Alloantibody responses were measured 2 weeks after transfusion, and cellular responses and growth factors were monitored over time. Results Contrary to our hypothesis, we found that chemotherapy led to increased alloantibody responses to allogeneic platelet transfusion. This enhanced response was antigen-specific and was associated with increased CD4+ and CD8+ T cell responses. Chemotherapy led to rapid lymphocyte depletion followed by reconstitution, non-specific activation of transitional B cells with the highest levels of activation in the least mature subsets, and increased serum levels of B cell activating factor (BAFF). Conclusion These data suggest that ablative chemotherapy can increase the risk of alloimmunization and, if confirmed clinically, that additional measures to protect these patient populations may be warranted.
Collapse
Affiliation(s)
- Rachael P. Jackman
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Orsolya Darst
- Vitalant Research Institute, San Francisco, CA, United States
| | - Betty Gaillard
- Vitalant Research Institute, San Francisco, CA, United States
| | - Johnson Q. Tran
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Mary M. Tomayko
- Department of Dermatology, Yale School of Medicine, New Haven, CT, United States
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States
| | - Marcus O. Muench
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
42
|
He L, Zhong C, Chang H, Inman JL, Celniker SE, Ioakeim-Ioannidou M, Liu KX, Haas-Kogan D, MacDonald SM, Threadgill DW, Kogan SC, Mao JH, Snijders AM. Genetic architecture of the acute and persistent immune cell response after radiation exposure. CELL GENOMICS 2023; 3:100422. [PMID: 38020972 PMCID: PMC10667298 DOI: 10.1016/j.xgen.2023.100422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/19/2023] [Accepted: 09/12/2023] [Indexed: 12/01/2023]
Abstract
Hematologic toxicity is a common side effect of multimodal cancer therapy. Nearly all animal studies investigating the causes of radiotherapy-induced hematologic toxicity use inbred strains with limited genetic diversity and do not reflect the diverse responses observed in humans. We used the population-based Collaborative Cross (CC) mouse resource to investigate the genetic architecture of the acute and persistent immune response after radiation exposure by measuring 22 immune parameters in 1,720 CC mice representing 35 strains. We determined relative acute and persistent radiation resistance scores at the individual strain level considering contributions from all immune parameters. Genome-wide association analysis identified quantitative trait loci associated with baseline and radiation responses. A cross-species radiation resistance score predicted recurrence-free survival in medulloblastoma patients. We present a community resource of immune parameters and genome-wide association analyses before and after radiation exposure for future investigations of the contributions of host genetics on radiosensitivity.
Collapse
Affiliation(s)
- Li He
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430079, China
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Chenhan Zhong
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Hang Chang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Jamie L. Inman
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Susan E. Celniker
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Comparative Biochemistry Program, University of California Berkeley, Berkeley, CA 94720, USA
| | | | - Kevin X. Liu
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shannon M. MacDonald
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David W. Threadgill
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX 77843, USA
- Departments of Nutrition and Cell Biology and Genetics, Texas A&M University, College Station, TX 77843, USA
| | - Scott C. Kogan
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Comparative Biochemistry Program, University of California Berkeley, Berkeley, CA 94720, USA
| | - Antoine M. Snijders
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Comparative Biochemistry Program, University of California Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
43
|
Zhang J, Yang L, Li H, Chan JW, Lee EK, Liu M, Ma L, Liu Q, Jin JY, Fu P, Xu Z, Kong FM(S. Dosimetric Effect of Thymus and Thoracic Duct on Radiation-Induced Lymphopenia in Patients With Primary Lung Cancer Who Received Thoracic Radiation. Adv Radiat Oncol 2023; 8:101260. [PMID: 38047216 PMCID: PMC10692302 DOI: 10.1016/j.adro.2023.101260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 04/20/2023] [Indexed: 12/04/2023] Open
Abstract
Purpose Radiation-induced lymphopenia is a well-recognized factor for tumor control and survival in patients with cancer. This study aimed to determine the role of radiation dose to the thymus and thoracic duct on radiation-induced lymphopenia. Methods and Materials Patients with primary lung cancer treated with thoracic radiation therapy between May 2015 and February 2020 with whole blood count data were eligible. Clinical characteristics, including age, gender, histology, stage, chemotherapy regimen, radiation dosimetry, and absolute lymphocyte count (ALC) were collected. The thymus and thoracic duct were contoured by one investigator for consistency and checked by one senior physician. The primary endpoint was radiation-induced decrease in lymphocytes, defined as the difference in ALC (DALC) before and after radiation therapy. Results The data of a total of 116 consecutive patients were retrospectively retrieved. Significant correlations were found between DALC and several clinical factors. These factors include stage, chemotherapy or concurrent chemoradiation, biologically effective dose (BED), mean lung dose, mean body dose, effective dose to immune cells (EDIC), mean thymus dose (MTD), and mean thoracic duct dose (MTDD) (all P < .05). Ridge regression showed that DALC = 0.0063 × BED + 0.0172 × EDIC + 0.0002 × MTD + 0.0147 × MTDD + 0.2510 (overall P = .00025 and F = 5.85). The combination model has the highest area under the curve of 0.77 (P < .001) when fitting the logistic regression model on DALC categorized as binary endpoint. The sensitivity and specificity of the combined model were 89% and 58%, respectively. Conclusions This study demonstrated for the first time that radiation doses to the thymus and thoracic duct are strongly associated with radiation-induced lymphopenia patients with lung cancer. Further validation studies are needed to implement thymus and thoracic duct as organs at risk.
Collapse
Affiliation(s)
| | | | | | | | | | - Min Liu
- Department of Respiratory Medicine, Hongkong University-Shenzhen Hospital, Shenzhen, China
| | | | | | - Jian-Yue Jin
- Department of Radiation Oncology, University Hospitals/Seidman Cancer Center and Case Comprehensive Cancer Center, Mentor, Ohio
| | - Pingfu Fu
- Department of Radiation Oncology, University Hospitals/Seidman Cancer Center and Case Comprehensive Cancer Center, Mentor, Ohio
| | | | | |
Collapse
|
44
|
Greenlund L, Shanley R, Mulford K, Neil EC, Lawrence J, Arnold S, Olin M, Pluhar GE, Venteicher AS, Chen CC, Ferreira C, Reynolds M, Cho LC, Wilke C, Shoo BA, Yuan J, Dusenbery K, Kleinberg LR, Terezakis SA, Sloan L. Comparison of peripheral leukocyte parameters in patients receiving conventionally and hypofractionated radiotherapy schemes for the treatment of newly diagnosed glioblastoma. Front Immunol 2023; 14:1284118. [PMID: 38022656 PMCID: PMC10644882 DOI: 10.3389/fimmu.2023.1284118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Treatment for glioblastomas, aggressive and nearly uniformly fatal brain tumors, provide limited long-term success. Immunosuppression by myeloid cells in both the tumor microenvironment and systemic circulation are believed to contribute to this treatment resistance. Standard multi-modality therapy includes conventionally fractionated radiotherapy over 6 weeks; however, hypofractionated radiotherapy over 3 weeks or less may be appropriate for older patients or populations with poor performance status. Lymphocyte concentration changes have been reported in patients with glioblastoma; however, monocytes are likely a key cell type contributing to immunosuppression in glioblastoma. Peripheral monocyte concentration changes in patients receiving commonly employed radiation fractionation schemes are unknown. Methods To determine the effect of conventionally fractionated and hypofractionated radiotherapy on complete blood cell leukocyte parameters, retrospective longitudinal concentrations were compared prior to, during, and following standard chemoradiation treatment. Results This study is the first to report increased monocyte concentrations and decreased lymphocyte concentrations in patients treated with conventionally fractionated radiotherapy compared to hypofractionated radiotherapy. Discussion Understanding the impact of fractionation on peripheral blood leukocytes is important to inform selection of dose fractionation schemes for patients receiving radiotherapy.
Collapse
Affiliation(s)
- Lindsey Greenlund
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Ryan Shanley
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Kellen Mulford
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Elizabeth C. Neil
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Jessica Lawrence
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Susan Arnold
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Michael Olin
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - G. Elizabeth Pluhar
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Andrew S. Venteicher
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Clark C. Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Clara Ferreira
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Margaret Reynolds
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - L. Chinsoo Cho
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Christopher Wilke
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - B. Aika Shoo
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Jianling Yuan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Kathryn Dusenbery
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Stephanie A. Terezakis
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Lindsey Sloan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
45
|
Laurent PA, Deutsch É. [Radiation-induced lymphopenia: Lymphocytes as a new organ at risk]. Cancer Radiother 2023; 27:511-518. [PMID: 37661506 DOI: 10.1016/j.canrad.2023.06.017] [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: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 09/05/2023]
Abstract
Taking the immune system into account in the fight against tumors has upset the cancer treatment paradigm in the 21st century. Combination treatment strategies associating radiotherapy with immunotherapy are being increasingly implemented in clinical practice. In this context, lymphocytes, whether lymphocytes infiltrating the tumour, circulating blood lymphocytes or lymphocytes residing within the lymph nodes, are key players in cellular and humoral anti-tumor immunity. The significant radiosensitivity of lymphocytes was demonstrated in the early 1990s. Along with the cells of the digestive mucosa, lymphocytes are thus among the most radiosensitive cell types in the body. Compared to the old practices of external radiotherapy, current intensity modulated treatments have allowed a considerable improvement in acute and late toxicity, at the cost of a significant increase in the volume irradiated at low doses. This is not without consequence on the incidence of radiation-induced lymphopenia, with prognostic implications for many tumor types. Thus, in order not to hinder the action of antitumor immunity and the efficacy of immunotherapy, it is essential to consider lymphocytes as a new organ at risk in its own right. In this development, based on current data from the literature, we will begin by justifying the necessary prevention of radiation-induced lymphopenia, before providing the tools currently known to apprehend lymphocytes as a new multicompartments. Finally, we will broaden the perspective by outlining ways to develop research in this area.
Collapse
Affiliation(s)
- P A Laurent
- Service de radiothérapie oncologique, Gustave-Roussy Cancer Campus, Villejuif, France; Inserm, U1030 Molecular Radiation Therapy and Therapeutic Innovation, Gustave-Roussy Cancer Campus, université Paris-Saclay, Villejuif, France
| | - É Deutsch
- Service de radiothérapie oncologique, Gustave-Roussy Cancer Campus, Villejuif, France; Inserm, U1030 Molecular Radiation Therapy and Therapeutic Innovation, Gustave-Roussy Cancer Campus, université Paris-Saclay, Villejuif, France.
| |
Collapse
|
46
|
Kut C, Midthune D, Lee E, Fair P, Cheunkarndee T, McNutt T, DeWeese T, Fakhry C, Kipnis V, Quon H. Developing the POTOMAC Model: A Novel Prediction Model to Study the Impact of Lymphopenia Kinetics on Survival Outcomes in Head and Neck Cancer Via an Ensemble Tree-Based Machine Learning Approach. JCO Clin Cancer Inform 2023; 7:e2300058. [PMID: 38096467 PMCID: PMC10735077 DOI: 10.1200/cci.23.00058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/25/2023] [Accepted: 10/19/2023] [Indexed: 12/18/2023] Open
Abstract
PURPOSE Lymphopenia is associated with poor survival outcomes in head and neck squamous cell carcinoma (HNSCC), yet there is no consensus on whether we should limit lymphopenia risks during treatment. To fully elucidate the prognostic role of baseline versus treatment-related lymphopenia, a robust analysis is necessary to investigate the relative importance of various lymphopenia metrics (LMs) in predicting survival outcomes. METHODS In this prospective cohort study, 363 patients were eligible for analysis (patients with newly diagnosed, nonmetastatic HNSCC treated with neck radiation with or without chemotherapy in 2015-2019). Data were acquired on 28 covariates: seven baseline, five disease, seven treatment, and nine LMs, including static and time-varying features for absolute lymphocyte count (ALC), neutrophil-to-lymphocyte ratio, and immature granulocytes (IGs). IGs were included, given their hypothesized role in inhibiting lymphocyte function. Overall, there were 4.0% missing data. Median follow-up was 2.9 years. We developed a model (POTOMAC) to predict survival outcomes using a random survival forest (RSF) procedure. RSF uses an ensemble approach to reduce the risk of overfitting and provides internal validation of the model using data that are not used in model development. The ability to predict survival risk was assessed using the AUC for the predicted risk score. RESULTS POTOMAC predicted 2-year survival with AUCs at 0.78 for overall survival (primary end point) and 0.73 for progression-free survival (secondary end point). Top modifiable risk factors included radiation dose and max ALC decrease. Top baseline risk factors included age, Charlson Comorbidity Index, Karnofsky Performance Score, and baseline IGs. Top-ranking LMs had superior prognostic performance when compared with human papillomavirus status, chemotherapy type, and dose (up to 2, 8, and 65 times higher in variable importance score). CONCLUSION POTOMAC provides important insights into potential approaches to reduce mortality in patients with HNSCC treated by chemoradiation but needs to be validated in future studies.
Collapse
Affiliation(s)
- Carmen Kut
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Doug Midthune
- Biometric Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Emerson Lee
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Peyton Fair
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Tia Cheunkarndee
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Todd McNutt
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Theodore DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Carole Fakhry
- Department of Otolaryngology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Victor Kipnis
- Biometric Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Harry Quon
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Otolaryngology, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
47
|
Paganetti H. A review on lymphocyte radiosensitivity and its impact on radiotherapy. Front Oncol 2023; 13:1201500. [PMID: 37601664 PMCID: PMC10435323 DOI: 10.3389/fonc.2023.1201500] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
It is well known that radiation therapy causes lymphopenia in patients and that this is correlated with a negative outcome. The mechanism is not well understood because radiation can have both immunostimulatory and immunosuppressive effects. How tumor dose conformation, dose fractionation, and selective lymph node irradiation in radiation therapy does affect lymphopenia and immune response is an active area of research. In addition, understanding the impact of radiation on the immune system is important for the design and interpretation of clinical trials combining radiation with immune checkpoint inhibitors, both in terms of radiation dose and treatment schedules. Although only a few percent of the total lymphocyte population are circulating, it has been speculated that their increased radiosensitivity may contribute to, or even be the primary cause of, lymphopenia. This review summarizes published data on lymphocyte radiosensitivity based on human, small animal, and in vitro studies. The data indicate differences in radiosensitivity among lymphocyte subpopulations that affect their relative contribution and thus the dynamics of the immune response. In general, B cells appear to be more radiosensitive than T cells and NK cells appear to be the most resistant. However, the reported dose-response data suggest that in the context of lymphopenia in patients, aspects other than cell death must also be considered. Not only absolute lymphocyte counts, but also lymphocyte diversity and activity are likely to be affected by radiation. Taken together, the reviewed data suggest that it is unlikely that radiation-induced cell death in lymphocytes is the sole factor in radiation-induced lymphopenia.
Collapse
Affiliation(s)
- Harald Paganetti
- Department of Radiation Oncology, Massachusetts General Hospital, Boston MA, United States
- Harvard Medical School, Boston MA, United States
| |
Collapse
|
48
|
Cai S, Fan Y, Guo Q, Sun Y, Zhao P, Tian Y, Fan Q. Impact of Radiation Dose to Circulating Immune Cells on Tumor Control and Survival in Esophageal Cancer. Cancer Biother Radiopharm 2023; 38:380-387. [PMID: 34883023 DOI: 10.1089/cbr.2021.0250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background: The immune system is well known to exert tumor immunosurveillance effects, and that immune cells circulating in the peripheral blood affect tumor prognosis. The study investigated the effect of estimated dose of radiation on circulating immune cells (EDRIC) and tumor control for esophageal cancer patients treated with concurrent chemo-radiotherapy. Materials and Methods: A total of 146 esophageal cancer patients treated with radiotherapy between January 2016 and June 2020 were retrospectively identified. We determined EDRIC, known prognostic factors, and the association of these factors with progression-free survival (PFS) and overall survival (OS). Results: The median follow-up was 17.9 months (2.7-60.4 months). The 3-year OS was 39.2%. Severe post-treatment lymphopenia was observed in 84.2% of patients. A negative correlation between EDRIC and absolute lymphocyte count was found (r = -0.679; p < 0.001). Patients with EDRIC ≥10.3 Gy were more likely to demonstrate grade 4 lymphopenia (55.2% vs. 4.5%; p < 0.001). Patients with grade 4 lymphopenia had a worse OS and PFS. On multivariate analysis, EDRIC was independently associated with OS (hazard ratio [HR], 1.142; p = 0.016) and PFS (HR, 1.121; p = 0.019). Conclusions: EDRIC can predict lymphocyte reduction and poor prognosis for esophageal cancer patients treated with radiotherapy.
Collapse
Affiliation(s)
- Shang Cai
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiation Oncology, Soochow University, Suzhou, China
| | - Yawen Fan
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiation Oncology, Soochow University, Suzhou, China
| | - Qi Guo
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiation Oncology, Soochow University, Suzhou, China
| | - Yanze Sun
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiation Oncology, Soochow University, Suzhou, China
| | - Peifeng Zhao
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiation Oncology, Soochow University, Suzhou, China
| | - Ye Tian
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiation Oncology, Soochow University, Suzhou, China
| | - Qiuhong Fan
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiation Oncology, Soochow University, Suzhou, China
| |
Collapse
|
49
|
Sharon S, Daher-Ghanem N, Zaid D, Gough MJ, Kravchenko-Balasha N. The immunogenic radiation and new players in immunotherapy and targeted therapy for head and neck cancer. FRONTIERS IN ORAL HEALTH 2023; 4:1180869. [PMID: 37496754 PMCID: PMC10366623 DOI: 10.3389/froh.2023.1180869] [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: 03/06/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023] Open
Abstract
Although treatment modalities for head and neck cancer have evolved considerably over the past decades, survival rates have plateaued. The treatment options remained limited to definitive surgery, surgery followed by fractionated radiotherapy with optional chemotherapy, and a definitive combination of fractionated radiotherapy and chemotherapy. Lately, immunotherapy has been introduced as the fourth modality of treatment, mainly administered as a single checkpoint inhibitor for recurrent or metastatic disease. While other regimens and combinations of immunotherapy and targeted therapy are being tested in clinical trials, adapting the appropriate regimens to patients and predicting their outcomes have yet to reach the clinical setting. Radiotherapy is mainly regarded as a means to target cancer cells while minimizing the unwanted peripheral effect. Radiotherapy regimens and fractionation are designed to serve this purpose, while the systemic effect of radiation on the immune response is rarely considered a factor while designing treatment. To bridge this gap, this review will highlight the effect of radiotherapy on the tumor microenvironment locally, and the immune response systemically. We will review the methodology to identify potential targets for therapy in the tumor microenvironment and the scientific basis for combining targeted therapy and radiotherapy. We will describe a current experience in preclinical models to test these combinations and propose how challenges in this realm may be faced. We will review new players in targeted therapy and their utilization to drive immunogenic response against head and neck cancer. We will outline the factors contributing to head and neck cancer heterogeneity and their effect on the response to radiotherapy. We will review in-silico methods to decipher intertumoral and intratumoral heterogeneity and how these algorithms can predict treatment outcomes. We propose that (a) the sequence of surgery, radiotherapy, chemotherapy, and targeted therapy should be designed not only to annul cancer directly, but to prime the immune response. (b) Fractionation of radiotherapy and the extent of the irradiated field should facilitate systemic immunity to develop. (c) New players in targeted therapy should be evaluated in translational studies toward clinical trials. (d) Head and neck cancer treatment should be personalized according to patients and tumor-specific factors.
Collapse
Affiliation(s)
- Shay Sharon
- Department of Oral and Maxillofacial Surgery, Hadassah Medical Center, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Oral and Maxillofacial Surgery, Boston University and Boston Medical Center, Boston, MA, United States
| | - Narmeen Daher-Ghanem
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Deema Zaid
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael J. Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
50
|
Makita K, Hamamoto Y, Kanzaki H, Nagasaki K, Kozuki T. Local control of bone metastasis treated with palliative radiotherapy in patients with lung cancer: An observational retrospective cohort study. Oncol Lett 2023; 26:303. [PMID: 37323814 PMCID: PMC10265332 DOI: 10.3892/ol.2023.13889] [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: 03/04/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Bone metastasis is common in advanced lung cancer, with the incidence reported to be 30%, and radiotherapy (RT) is used for pain relief from bone metastasis. The present study aimed to identify factors affecting local control (LC) of bone metastasis from lung cancer and to assess the significance of moderate RT dose escalation. This was a retrospective cohort study, where LC of bone metastasis from lung cancer that had received palliative RT was reviewed. LC at RT sites was evaluated with follow-up computed tomography (CT). The influence of treatment-, cancer- and patient-related risk factors for LC was assessed. A total of 317 metastatic lesions in 210 patients with lung cancer were evaluated. The median RT dose (biologically effective dose calculated using an α/β of 10 Gy; BED10) was 39.0 Gy (range, 14.4-50.7 Gy). The median follow-up time for survival and median radiographic follow-up time were 8 (range, 1-127) and 4 (range, 1-124) months, respectively. The 0.5-year overall survival and LC rates were 58.9 and 87.7%, respectively. The local recurrence rate in RT sites was 11.0%, and bone metastatic progression, except in RT sites, was observed in 46.1% at the time of local recurrence or the last follow-up CT of the RT sites. According to multivariate analysis, RT sites, pre-RT neutrophil to lymphocyte ratio (NLR), post-RT non-administration of molecular-targeting agents (MTs), and non-administration of bone modifying agents (BMAs) were significant unfavorable factors for LC of bone metastasis. Moderate RT dose escalation (BED10 >39 Gy) tended to improve the LC of RT sites. In cases without MTs, moderate dose escalation of RT dose improved the LC of RT sites. In conclusion, treatment (post-RT MTs and BMAs), cancer (RT sites) and patient (pre-RT NLR)-related risk factors had a large impact on improving the LC of RT sites. Moderate RT dose escalation seemed to have a small impact on improving the LC of RT sites.
Collapse
Affiliation(s)
- Kenji Makita
- Department of Radiation Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime 791-0280, Japan
- Department of Radiology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
- Department of Radiology, Ehime Prefectural Central Hospital, Matsuyama, Ehime 790-0024, Japan
| | - Yasushi Hamamoto
- Department of Radiation Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime 791-0280, Japan
| | - Hiromitsu Kanzaki
- Department of Radiation Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime 791-0280, Japan
| | - Kei Nagasaki
- Department of Radiation Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime 791-0280, Japan
| | - Toshiyuki Kozuki
- Department of Thoracic Oncology and Medicine, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime 791-0280, Japan
| |
Collapse
|