1
|
Xing Y, Moore C, Saha D, Nguyen D, Bleile M, Jia X, Timmerman R, Peng H, Jiang S. Mathematical modeling of the synergetic effect between radiotherapy and immunotherapy. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2025; 22:1206-1225. [PMID: 40296809 DOI: 10.3934/mbe.2025044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The synergy between radiotherapy and immunotherapy plays a pivotal role in enhancing tumor control and treatment outcomes. To explore the underlying mechanisms of synergy, we investigated a novel treatment approach known as personalized ultra-fractionated stereotactic adaptive radiation (PULSAR) therapy, which emphasizes the impact of radiation timing. Unlike conventional daily treatments, PULSAR delivers high-dose radiation in spaced intervals over weeks or months, enabling tumors to adapt and potentially enhancing synergy with immunotherapy. Drawing on insights from small-animal radiation studies, we developed a discrete-time model based on multiple difference equations to elucidate the temporal dynamics of tumor control driven by both radiation and the adaptive immune response. By accounting for the migration and infiltration of T cells within the tumor microenvironment, we established a quantitative link between radiation therapy and immunotherapy. Model parameters were estimated using a simulated annealing algorithm applied to training data, and our model achieved high accuracy for the test data with a root mean square error of 287 mm3. Notably, our framework replicated the PULSAR effect observed in animal studies, revealing that longer intervals between radiation treatments in the context of immunotherapy yielded enhanced tumor control. Specifically, mice receiving immunotherapy alongside radiation pulses delivered at extended intervals, ten days, showed markedly improved tumor responses, whereas those treated with shorter intervals did not achieve comparable benefits. Moreover, our model offers an in-silico tool for designing future personalized ultra-fractionated stereotactic adaptive radiation trials. Overall, these findings underscore the critical importance of treatment timing in harnessing the synergy between radiotherapy and immunotherapy and highlight the potential of our model to optimize and individualize treatment protocols.
Collapse
Affiliation(s)
- Yixun Xing
- Medical Artificial Intelligence and Automation Laboratory, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Advanced Data Analytics, University of North Texas, Denton, TX 76205, USA
| | - Casey Moore
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Debabrata Saha
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Dan Nguyen
- Medical Artificial Intelligence and Automation Laboratory, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - MaryLena Bleile
- Medical Artificial Intelligence and Automation Laboratory, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Statistical Science, Southern Methodist University, Dallas, TX 75275, USA
| | - Xun Jia
- Medical Artificial Intelligence and Automation Laboratory, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Robert Timmerman
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hao Peng
- Medical Artificial Intelligence and Automation Laboratory, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Steve Jiang
- Medical Artificial Intelligence and Automation Laboratory, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
2
|
Zhou S, Yang H. Radiotherapy modulates autophagy to reshape the tumor immune microenvironment to enhance anti-tumor immunity in esophageal cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189302. [PMID: 40120778 DOI: 10.1016/j.bbcan.2025.189302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/15/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
The combination of radiotherapy and immunotherapy exerts synergistic antitumor in a range of human cancers, and also in esophageal cancer. Radiotherapy-induced tumor immune microenvironment (TIME) reprogramming is an essential basis for the synergistic antitumor between radiotherapy and immunotherapy. Radiotherapy can induce autophagy in tumor cells and immune cells of TIME, and autophagy activation is involved in the modification of immunological characteristics of TIME. The TIME landscape of esophageal cancer, especially ESCC, can be affected by radiotherapy or autophagy regulation. In this review, we depicted that local radiotherapy-induced autophagy could promote the maturation, migration, infiltration, and function of immune cells by complicated mechanisms to make TIME from immune "cold" to "hot", resulting in the synergistic antitumor of RT and IO. We argue that unraveling the relevance of radiotherapy-initiated autophagy to driving radiotherapy reprogramming TIME will open new ideas to explore new targets or more efficiently multimodal therapeutic interventions in ESCC.
Collapse
Affiliation(s)
- Suna Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Department of Radiation Oncology, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China
| | - Haihua Yang
- Key Laboratory of Radiation Oncology of Taizhou, Department of Radiation Oncology, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China.
| |
Collapse
|
3
|
Cheng SH, Tu KY, Lee HH. The dynamic duo: A narrative review on the synergy between stereotactic body radiotherapy and immunotherapy in lung cancer treatment (Review). Oncol Rep 2024; 52:96. [PMID: 38874014 PMCID: PMC11188053 DOI: 10.3892/or.2024.8755] [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: 01/08/2024] [Accepted: 05/10/2024] [Indexed: 06/15/2024] Open
Abstract
Immunotherapy, particularly immune checkpoint inhibitors (ICIs), is undoubtedly one of the major breakthroughs in lung cancer research. Patient survival and prognosis have all been improved as a result, although numerous patients do not respond to immunotherapy due to various immune escape mechanisms of the tumor cells. Recent preclinical and clinical evidence has shown that stereotactic body radiotherapy (SBRT), also known as stereotactic ablative radiotherapy, has a prominent immune priming effect that could elicit antitumor immunity against specific tumor antigens and destroy distant tumor cells, thereby achieving the elusive abscopal effect, with the resulting immuno‑active tumor environment also being more conducive to ICIs. Some landmark trials have already demonstrated the survival benefit of the dynamic duo of SBRT plus immunotherapy in metastatic non‑small‑cell lung cancer, while others such as PEMBRO‑RT further suggest that the addition of SBRT to immunotherapy could expand the current indication to those who have historically responded poorly to ICIs. In the present review, the biological mechanisms that drive the synergistic effect of SBRT and immunotherapy were first briefly outlined; then, the current understanding from clinical trials was summarized and new insight into the evolving role of immunotherapy and SBRT synergy in lung cancer treatment was provided. Finally, novel avenues for discovery were highlighted. The innovation of the present review lies in the inclusion of non‑ICI immunotherapy in the discussion, which provides a more comprehensive view on the current development and future trend of SBRT + immunotherapy synergy.
Collapse
Affiliation(s)
- Sarah Hsin Cheng
- Department of Clinical Education and Training, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Kuan-Yi Tu
- School of Post Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Hsin-Hua Lee
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
- PhD Program in Environmental and Occupational Medicine, Kaohsiung Medical University and National Health Research Institutes, Kaohsiung 807, Taiwan, R.O.C
- Department of Radiation Oncology, Faculty of Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
4
|
Choudhury AD, Kwak L, Cheung A, Allaire KM, Marquez J, Yang DD, Tripathi A, Kilar JM, Flynn M, Maynard B, Reichel R, Pace AF, Chen BK, Van Allen EM, Kilbridge K, Wei XX, McGregor BA, Pomerantz MM, Bhatt RS, Sweeney CJ, Bubley GJ, Jacene HA, Taplin ME, Huang FW, Harshman LC, Fong L. Randomized Phase II Study Evaluating the Addition of Pembrolizumab to Radium-223 in Metastatic Castration-resistant Prostate Cancer. Cancer Immunol Res 2024; 12:704-718. [PMID: 38552171 PMCID: PMC11148544 DOI: 10.1158/2326-6066.cir-22-0306] [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: 04/26/2022] [Revised: 08/15/2023] [Accepted: 03/08/2024] [Indexed: 06/05/2024]
Abstract
The checkpoint immunotherapeutic pembrolizumab induces responses in a small minority of patients with metastatic castration-resistant prostate cancer (mCRPC). Radium-223 (R223) may increase immunogenicity of bone metastases and increase pembrolizumab (P) activity. In a randomized phase II study, we assessed the effect of R223+P compared with R223 on tumor immune infiltration, safety, and clinical outcomes in patients with mCRPC. The primary endpoint was differences in CD4+ and CD8+ T-cell infiltrate in 8-week versus baseline bone metastasis biopsies; secondary endpoints were safety, radiographic progression-free survival (rPFS), and overall survival (OS). Of the 42 treated patients (29 R223+P, 13 R223), 18 R223+P and 8 R223 patients had evaluable paired tumor biopsies. Median fold-change of CD4+ T cells was -0.7 (range: -9.3 to 4.7) with R223+P and 0.1 (-11.1 to 3.7) with R223 (P = 0.66); for CD8+ T cells, median fold-change was -0.6 (-7.4 to 5.3) with R223+P and -1.3 (-3.1 to 4.8) with R223 (P = 0.66). Median rPFS and OS was 6.1 (95% confidence interval: 2.7-11.0) and 16.9 months [12.7-not reached (NR)], respectively, with R223+P and 5.7 (2.6-NR) and 16.0 (9.0-NR), respectively, with R223. Although R223+P was well tolerated with no unexpected toxicity, the combination did not improve efficacy. High-dimensional flow cytometry demonstrated minimal immune modulation with R223, whereas R223+P induced CTLA-4 expression on circulating CD4+ T cells. Clinical responders possessed lower circulating frequencies of Ki67+ T and myeloid cells at baseline and higher circulating frequencies of TIM-3+ T and myeloid cells by week 9. Although R223+P did not induce T-cell infiltration into the tumor microenvironment, exhaustion of induced peripheral T-cell immune responses may dampen the combination's clinical activity.
Collapse
Affiliation(s)
- Atish D. Choudhury
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Lucia Kwak
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alexander Cheung
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Kathryn M. Allaire
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Jaqueline Marquez
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - David D. Yang
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | - Rebecca Reichel
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | | | - Brandon K. Chen
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Eliezer M. Van Allen
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Kerry Kilbridge
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Xiao X. Wei
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Bradley A. McGregor
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Mark M. Pomerantz
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Rupal S. Bhatt
- Harvard Medical School, Boston, Massachusetts
- Beth-Israel Deaconess Medical Center, Boston, Massachusetts
| | | | - Glenn J. Bubley
- Harvard Medical School, Boston, Massachusetts
- Beth-Israel Deaconess Medical Center, Boston, Massachusetts
| | - Heather A. Jacene
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Mary-Ellen Taplin
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Franklin W. Huang
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | | | - Lawrence Fong
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| |
Collapse
|
5
|
Castelluccia A, Sardaro A, Niccoli Asabella A, Pisani AR, Rubini D, Portaluri M, Tramacere F. Durable complete response to PET-CT driven stereotactic radiation therapy plus pembrolizumab for pleomorphic Pancoast cancer: Case report and literature review. Clin Case Rep 2024; 12:e8633. [PMID: 38585585 PMCID: PMC10996042 DOI: 10.1002/ccr3.8633] [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: 12/27/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/09/2024] Open
Abstract
PET-driven SBRT plus pembrolizumab as first-line therapy against pleomorphic Pancoast cancer appears beneficial, probably due to high equivalent doses of SBRT on photopenic necrotic core and synergic immune system stimulation of immunoradiotherapy.
Collapse
Affiliation(s)
| | - Angela Sardaro
- Section of Radiology and Radiation Oncology, Interdisciplinary Department of MedicineUniversity of Bari “Aldo Moro”BariItaly
| | - Artor Niccoli Asabella
- Section of Nuclear Medicine, Interdisciplinary Department of MedicineUniversity of Bari Aldo MoroBariItaly
| | - Antonio Rosario Pisani
- Section of Nuclear Medicine, Interdisciplinary Department of MedicineUniversity of Bari Aldo MoroBariItaly
| | - Dino Rubini
- Department of Precision MedicineUniversità degli Studi della Campania Luigi VanvitelliNapoliCampaniaItaly
| | | | | |
Collapse
|
6
|
Swamy K. Therapeutic In Situ Cancer Vaccine Using Pulsed Stereotactic Body Radiotherapy-A Translational Model. Vaccines (Basel) 2023; 12:7. [PMID: 38276666 PMCID: PMC10819354 DOI: 10.3390/vaccines12010007] [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: 10/24/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Both radiation and cancer therapeutic vaccine research are more than 100 years old, and their potential is likely underexplored. Antiangiogenics, nanoparticle targeting, and immune modulators are some other established anticancer therapies. In the meantime, immunotherapy usage is gaining momentum in clinical applications. This article proposes the concept of a pulsed/intermittent/cyclical endothelial-sparing single-dose in situ vaccination (ISVRT) schedule distinguishable from the standard therapeutic stereotactic body radiotherapy (SBRT) and stereotactic radiosurgery (SRS) plans. This ISVRT schedule can repeatedly generate tumor-specific neoantigens and epitopes for primary and immune modulation effects, augment supplementary immune enhancement techniques, activate long-term memory cells, avoid extracellular matrix fibrosis, and essentially synchronize with the vascular normalized immunity cycle. The core mechanisms of ISVRT impacting in situ vaccination would be optimizing cascading antigenicity and adjuvanticity. The present proposed hypothesis can be validated using the algorithm presented. The indications for the proposed concept are locally progressing/metastatic cancers that have failed standard therapies. Immunotherapy/targeted therapy, chemotherapy, antiangiogenics, and vascular-lymphatic normalization are integral to such an approach.
Collapse
|
7
|
Huang L, Yin Y, Qian D, Cao Y, Wang D, Wu X, Ming L, Huang Z, Zhou L. IRF7 and IFIT2 in mediating different hemorrhage outcomes for non-small cell lung cancer after bevacizumab treatment. J Thorac Dis 2023; 15:2022-2036. [PMID: 37197507 PMCID: PMC10183489 DOI: 10.21037/jtd-23-389] [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: 02/15/2023] [Accepted: 04/20/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Lung cancer has some of the highest morbidity and mortality worldwide among cancers, with non-small cell lung cancer (NSCLC) accounting for 85% of lung cancer diagnoses. Severe pulmonary hemorrhage (PH) is a serious potential adverse event in the treatment of lung cancer with bevacizumab. Significant clinical differences have been observed between patients with lung adenocarcinoma (LUAD) and those with lung squamous cell carcinoma (LUSC) after bevacizumab treatment; however, the underlying causes is unclear and requires further study. METHODS First, tumor tissues from LUAD and LUSC patients were stained with antibodies targeting CD31 and CD34 to assess the difference in microvessel density (MVD). Tube formation assays were performed using HMEC-1 cells cocultured with lung cancer cells. Single-cell sequencing data obtained from lung cancer tissues were then downloaded and analyzed to identify differentially expressed genes related to angiogenesis in LUAD and LUSC tumors. Real-time polymerase chain reaction, immunofluorescence analysis, small interfering RNA analysis, and enzyme-linked immunosorbent assay were performed to clarify the underlying causes. RESULTS The MVD of LUAD tissues was higher than that of LUSC tissues. Additionally, endothelial cells cocultured with LUAD cells had a higher MVD than did those cocultured with LUSC cells. Although bevacizumab mainly targets vascular endothelial growth factor (VEGF), the expression of VEGF in LUSC and LUAD cells was not significantly different (P>0.05). Further experiments showed that interferon regulatory factor 7 (IRF7) and interferoninduced protein with tetratricopeptide repeats 2 (IFIT2) were differentially expressed between LUSC and LUAD tumors. Higher IRF7 levels and lower IFIT2 levels in LUAD tumors were associated with higher MVD in LUAD tissues, which may be responsible for the different hemorrhage outcomes after bevacizumab treatment. CONCLUSIONS Our data indicated that IRF7 and IFIT2 may account for the differential hemorrhage outcomes in patients with NSCLC after bevacizumab treatment, revealing a new mechanism underlying bevacizumab-induced pulmonary hemoptysis.
Collapse
Affiliation(s)
- Liuying Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yuan Yin
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Danqi Qian
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yulin Cao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Duo Wang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaohan Wu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Liang Ming
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Leyuan Zhou
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
- Department of Radiation Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
8
|
Liu C, Wang X, Qin W, Tu J, Li C, Zhao W, Ma L, Liu B, Qiu H, Yuan X. Combining radiation and the ATR inhibitor berzosertib activates STING signaling and enhances immunotherapy via inhibiting SHP1 function in colorectal cancer. Cancer Commun (Lond) 2023; 43:435-454. [PMID: 36855844 PMCID: PMC10091106 DOI: 10.1002/cac2.12412] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/03/2023] [Accepted: 02/17/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) have shown a moderate response in colorectal cancer (CRC) with deficient mismatch repair (dMMR) functions and poor response in patients with proficient MMR (pMMR). pMMR tumors are generally immunogenically "cold", emphasizing combination strategies to turn the "cold" tumor "hot" to enhance the efficacy of ICIs. ATR inhibitors (ATRi) have been proven to cooperate with radiation to promote antitumor immunity, but it is unclear whether ATRi could facilitate the efficacy of IR and ICI combinations in CRCs. This study aimed to investigate the efficacy of combining ATRi, irradiation (IR), and anti-PD-L1 antibodies in CRC mouse models with different microsatellite statuses. METHODS The efficacy of combining ATRi, IR, and anti-PD-L1 antibodies was evaluated in CRC tumors. The tumor microenvironment and transcriptome signatures were investigated under different treatment regimens. The mechanisms were explored via cell viability assay, flow cytometry, immunofluorescence, immunoblotting, co-immunoprecipitation, and real-time quantitative PCR in multiple murine and human CRC cell lines. RESULTS Combining ATRi berzosertib and IR enhanced CD8+ T cell infiltration and enhanced the efficacy of anti-PD-L1 therapy in mouse CRC models with different microsatellite statuses. The mechanistic study demonstrated that IR + ATRi could activate both the canonical cGAS-STING-pTBK1/pIRF3 axis by increasing cytosolic double-stranded DNA levels and the non-canonical STING signaling by attenuating SHP1-mediated inhibition of the TRAF6-STING-p65 axis, via promoting SUMOylation of SHP1 at lysine 127. By boosting the STING signaling, IR + ATRi induced type I interferon-related gene expression and strong innate immune activation and reinvigorated the cold tumor microenvironment, thus facilitating immunotherapy. CONCLUSIONS The combination of ATRi and IR could facilitate anti-PD-L1 therapy by promoting STING signaling in CRC models with different microsatellite statuses. The new combination strategy raised by our study is worth investigating in the management of CRC.
Collapse
Affiliation(s)
- Chaofan Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xi Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Wan Qin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Chunya Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Li Ma
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Hong Qiu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| |
Collapse
|
9
|
Janopaul-Naylor JR, Cao Y, McCall NS, Switchenko JM, Tian S, Chen H, Stokes WA, Kesarwala AH, McDonald MW, Shelton JW, Bradley JD, Higgins KA. Definitive intensity modulated proton re-irradiation for lung cancer in the immunotherapy era. Front Oncol 2023; 12:1074675. [PMID: 36733369 PMCID: PMC9888533 DOI: 10.3389/fonc.2022.1074675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/29/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction As immunotherapy has improved distant metastasis-free survival (DMFS) in Non-Small Cell Lung Cancer (NSCLC), isolated locoregional recurrences have increased. However, management of locoregional recurrences can be challenging. We report our institutional experience with definitive intent re-irradiation using Intensity Modulated Proton Therapy (IMPT). Method Retrospective cohort study of recurrent or second primary NSCLC or LS-SCLC treated with IMPT. Kaplan-Meier method and log-rank test were used for time-to-event analyses. Results 22 patients were treated from 2019 to 2021. After first course of radiation (median 60 Gy, range 45-70 Gy), 45% received adjuvant immunotherapy. IMPT re-irradiation began a median of 28.2 months (8.8-172.9 months) after initial radiotherapy. The median IMPT dose was 60 GyE (44-60 GyE). 36% received concurrent chemotherapy with IMPT and 18% received immunotherapy after IMPT. The median patient's IMPT lung mean dose was 5.3 GyE (0.9-13.9 GyE) and 5 patients had cumulative esophagus max dose >100 GyE with 1-year overall survival (OS) 68%, 1-year local control 80%, 1-year progression free survival 45%, and 1-year DMFS 60%. Higher IMPT (HR 1.4; 95% CI 1.1-1.7, p=0.01) and initial radiotherapy mean lung doses (HR 1.3; 95% CI 1.0-1.6, p=0.04) were associated with worse OS. Two patients developed Grade 3 pneumonitis or dermatitis, one patient developed Grade 2 pneumonitis, and seven patients developed Grade 1 toxicity. There were no Grade 4 or 5 toxicities. Discussion Definitive IMPT re-irradiation for lung cancer can prolong disease control with limited toxicity, particularly in the immunotherapy era.
Collapse
Affiliation(s)
- James R. Janopaul-Naylor
- Winship Cancer Institute, Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Yichun Cao
- Biostatistics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Neal S. McCall
- Winship Cancer Institute, Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Jeffrey M. Switchenko
- Biostatistics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, United States
- Rollins School of Public Health, Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, United States
| | - Sibo Tian
- Winship Cancer Institute, Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Haijian Chen
- Winship Cancer Institute, Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - William A. Stokes
- Winship Cancer Institute, Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Aparna H. Kesarwala
- Winship Cancer Institute, Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Mark W. McDonald
- Winship Cancer Institute, Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Joseph W. Shelton
- Winship Cancer Institute, Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Jeffrey D. Bradley
- Winship Cancer Institute, Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Kristin A. Higgins
- Winship Cancer Institute, Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
10
|
Yang H, Miao Y, Yu Z, Wei M, Jiao X. Cell adhesion molecules and immunotherapy in advanced non-small cell lung cancer: Current process and potential application. Front Oncol 2023; 13:1107631. [PMID: 36895477 PMCID: PMC9989313 DOI: 10.3389/fonc.2023.1107631] [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: 11/25/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
Advanced non-small cell lung cancer (NSCLC) is a severe disease and still has high mortality rate after conventional treatment (e.g., surgical resection, chemotherapy, radiotherapy and targeted therapy). In NSCLC patients, cancer cells can induce immunosuppression, growth and metastasis by modulating cell adhesion molecules of both cancer cells and immune cells. Therefore, immunotherapy is increasingly concerned due to its promising anti-tumor effect and broader indication, which targets cell adhesion molecules to reverse the process. Among these therapies, immune checkpoint inhibitors (mainly anti-PD-(L)1 and anti-CTLA-4) are most successful and have been adapted as first or second line therapy in advanced NSCLC. However, drug resistance and immune-related adverse reactions restrict its further application. Further understanding of mechanism, adequate biomarkers and novel therapies are necessary to improve therapeutic effect and alleviate adverse effect.
Collapse
Affiliation(s)
- Hongjian Yang
- Innovative Institute, China Medical University, Shenyang, China
| | - Yuxi Miao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Centre, Shenyang, China
| | - Xue Jiao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Shenyang Kangwei Medical Laboratory Analysis Co. LTD, Shenyang, China
| |
Collapse
|
11
|
Stereotactic Body Radiotherapy and Immunotherapy for Older Patients with Oligometastases: A Proposed Paradigm by the International Geriatric Radiotherapy Group. Cancers (Basel) 2022; 15:cancers15010244. [PMID: 36612239 PMCID: PMC9818761 DOI: 10.3390/cancers15010244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
The standard of care for metastatic disease is systemic therapy. A unique subset of patients with limited metastatic disease defined as distant involvement of five anatomic sites or less (oligometastases) have a better chance of remission or improved survival and may benefit from local treatments such as surgery or stereotactic body radiotherapy (SBRT). However, to prevent further spread of disease, systemic treatment such as chemotherapy, targeted therapy, and hormonal therapy may be required. Older patients (70 years old or above) or physiologically frail younger patients with multiple co-morbidities may not be able to tolerate the conventional chemotherapy due to its toxicity. In addition, those with a good performance status may not receive optimal chemotherapy due to concern about toxicity. Recently, immunotherapy with checkpoint inhibitors (CPI) has become a promising approach only in the management of program death ligand 1 (PD-L1)-positive tumors. Thus, a treatment method that elicits induction of PD-L1 production by tumor cells may allow all patients with oligometastases to benefit from immunotherapy. In vitro studies have demonstrated that high dose of radiotherapy may induce formation of PD-L1 in various tumors as a defense mechanism against inflammatory T cells. Clinical studies also corroborated those observations. Thus, SBRT, with its high precision to minimize damage to normal organs, may be a potential treatment of choice for older patients with oligometastases due to its synergy with immunotherapy. We propose a protocol combining SBRT to achieve a minimum radiobiologic equivalent dose around 59.5 Gy to all tumor sites if feasible, followed four to six weeks later by CPI for those cancer patients with oligometastases. All patients will be screened with frailty screening questionnaires to identify individuals at high risk for toxicity. The patients will be managed with an interdisciplinary team which includes oncologists, geriatricians, nurses, nutritionists, patient navigators, and social workers to manage all aspects of geriatric patient care. The use of telemedicine by the team may facilitate patient monitoring during treatment and follow-up. Preliminary data on toxicity, local control, survival, and progression-free survival may be obtained and serve as a template for future prospective studies.
Collapse
|
12
|
Varlotto JM, Sun Z, Ky B, Upshaw J, Fitzgerald TJ, Diehn M, Lovly C, Belani C, Oettel K, Masters G, Harkenrider M, Ross H, Ramalingam S, Pennell NA. A Review of Concurrent Chemo/Radiation, Immunotherapy, Radiation Planning, and Biomarkers for Locally Advanced Non-small Cell Lung Cancer and Their Role in the Development of ECOG-ACRIN EA5181. Clin Lung Cancer 2022; 23:547-560. [PMID: 35882620 DOI: 10.1016/j.cllc.2022.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 01/27/2023]
Abstract
ECOG-ACRIN EA5181 is a current prospective, randomized trial that is investigating whether the addition of concomitant durvalumab to standard chemo/radiation followed by 1 year of consolidative durvalumab results in an overall survival benefit over standard chemo/radiation alone followed by 1 year of consolidative durvalumab in patients with locally advanced, unresectable non-small cell lung cancer (NSCLC). Because multiple phase I/II trials have shown the relative safety of adding immunotherapy to chemo/radiation and due to the known synergism between chemotherapy and immunotherapy, it is hoped that concomitant durvalumab can reduce the relatively high incidence of local failure (38%-46%) as seen in recent prospective, randomized trials of standard chemo/radiation in this patient population. We will review the history of radiation for LA-NSCLC and discuss the role of induction, concurrent and consolidative chemotherapy as well as the concerns for late cardiac and pulmonary toxicities associated with treatment. Furthermore, we will review the potential role of next generation sequencing, PD-L1, ctDNA and tumor mutation burden and their possible impact on this trial.
Collapse
Affiliation(s)
- John Michael Varlotto
- Department of Oncology, Edwards Comprehensive Cancer Center/Marshall University, Huntington, WV.
| | - Zhuoxin Sun
- Dana Farber Cancer Institute - ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Bonnie Ky
- Division of Cardiovascular Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jenica Upshaw
- Department of Medicine, Tufts University, Boston, MA
| | | | - Max Diehn
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Christine Lovly
- Division of Hematology Oncology, Vanderbilt University, Nashville, TN
| | - Chandra Belani
- Department of Medical Oncology, Penn State Cancer Institute, Hershey, PA
| | - Kurt Oettel
- Department of Medical Oncology, Gundersen Lutheran Medical Center, La Crosse, WI
| | | | - Matthew Harkenrider
- Department of Radiation Oncology, Stritch School of Medicine Loyola University Chicago, Maywood, IL
| | - Helen Ross
- Department of Medical Oncology, Banner MD Anderson Cancer Center, Gilbert, AZ
| | | | - Nathan A Pennell
- Department of Hematology Oncology, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
13
|
Immunotherapy and Radiotherapy as an Antitumoral Long-Range Weapon-A Partnership with Unsolved Challenges: Dose, Fractionation, Volumes, Therapeutic Sequence. Curr Oncol 2022; 29:7388-7395. [PMID: 36290857 PMCID: PMC9601214 DOI: 10.3390/curroncol29100580] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy, the modern oncological treatment with immune checkpoint inhibitors (ICIs), has been part of the clinical practice for malignant melanoma for more than a decade. Anti-cytotoxic T-lymphocyte antigen 4 (CTLA4), anti-programmed cell death Protein 1 (PD-1), or anti programmed death-ligand 1 (PD-L1) agents are currently part of the therapeutic arsenal of metastatic or relapsed disease in numerous cancers; more recently, they have also been evaluated and validated as consolidation therapy in the advanced local stage. The combination with radiotherapy, a treatment historically considered loco-regional, changes the paradigm, offering-via synergistic effects-the potential to increase immune-mediated tumor destruction. However, the fragile balance between the tumoricidal effects through immune mechanisms and the immunosuppression induced by radiotherapy means that, in the absence of ICI, the immune-mediated potentiation effect of radiotherapy at a distance from the site of administration is rare. Through analysis of the preclinical and clinical data, especially the evidence from the PACIFIC clinical trial, we can consider that hypofractionated irradiation and reduction of the irradiated volume, in order to protect the immune-infiltrated tumor microenvironment, performed concurrently with the immunotherapy or a maximum of 2 weeks before the start of ICI treatment, could bring maximum benefits. In addition, avoiding radiation-induced lymphopenia (RILD) by protecting some anatomical lymphoid structures or large blood vessels, as well as the use of irradiation of partial tumor volumes, even in plurimetastatic disease, for the conversion of a "cold" immunological tumor into a "hot" immunological tumor are modern concepts of radiotherapy in the era of immunotherapy. Low-dose radiotherapy could also be proposed in plurimetastatic cases, the effect being different (modeling of the TME) from that of high doses per fraction irradiation (cell death with release of antigens that facilitates immune-mediated cell death).
Collapse
|
14
|
Xiao Y, Zhuang H. Effect of stereotactic radiotherapy on immune microenvironment of lung cancer. Front Immunol 2022; 13:1025872. [PMID: 36211382 PMCID: PMC9540518 DOI: 10.3389/fimmu.2022.1025872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/02/2022] [Indexed: 12/05/2022] Open
Abstract
Stereotactic radiotherapy (SRT) is one of the main treatment modalities for lung cancer, and the current SRT approach combined with immunotherapy has initially presented good clinical efficacy in lung cancer. SRT activates the immune system through in situ immunization, releasing antigens into the blood, which promotes the antigen–antibody response and then induces tumor cell apoptosis. Dose fractionation has different effects on the immune microenvironment, and the tumor microenvironment after SRT also changes over time, all of which have an impact on SRT combined immunotherapy. Although much research on the immune microenvironment of SRT has been conducted, many problems still require further exploration.
Collapse
|
15
|
Yao N, Qin Z, Ma J, Lu J, Sun K, Zhang Y, Qu W, Cui L, Yuan S, Jiang A, Li N, Tong S, Yao Y. Anti-programmed death ligand 1 immunotherapy in patients with limited-stage small cell lung cancer: a real-world exploratory study. J Chemother 2022:1-7. [PMID: 36124811 DOI: 10.1080/1120009x.2022.2125750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
The aim of this study was to investigate the safety and efficacy of anti-programmed death ligand 1 (PD-L1) immunotherapy plus chemoradiotherapy for patients with limited-stage small cell lung cancer (LS-SCLC) in clinical practice. Patients with LS-SCLC treated with anti-PD-L1 (atezolizumab/durvalumab) plus chemoradiotherapy (CRT) as the initial treatment at three general hospitals between March 2020 and December 2021 were retrospectively analysed. 1:2 propensity score matching for controls that receive CRT only was performed. Clinical data (age, sex, history of cancer treatment, adverse events, etc.) were collected to evaluate toxicity, progression-free survival (PFS) and objective response rate (ORR). Researchers used univariate Chi-squared analyses to determine if anti-PD-L1 immunotherapy had a significant association with toxicity or ORR. Kaplan-Meier survival analysis, and the log-rank test were used to compare survival curves between the two groups. In the anti-PD-L1 plus CRT and CRT groups, 15 and 30 patients were analyzed; median follow-up was 16.39 months and 16.64 months, respectively. Incidence of toxicity between the two groups was similar and there were no new safety signals. Anti-PD-L1 immunotherapy significantly improved PFS (P = 0.02). The median PFS was not reached in the anti-PD-L1 plus CRT group versus 8.18 months [95% confidence interval (CI), 6.14-10.22 months] in the CRT group. The ORR were 93.33% and 76.67%, respectively (P = 0.34). This study supports adding anti-PD-L1 immunotherapy (atezolizumab/durvalumab) to CRT as an initial treatment option in patients with LS-SCLC for its favorable safety profile and efficacy.
Collapse
Affiliation(s)
- Nan Yao
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhaohui Qin
- Research Center for Medical and Health Emergency Rescue, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ji Ma
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiaying Lu
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kaiguo Sun
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yiqing Zhang
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wanxi Qu
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Li Cui
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shiwang Yuan
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Aijun Jiang
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Na Li
- Department of Radiation Oncology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Shaodong Tong
- Department of Radiation Oncology, The Third People's Hospital of Xuzhou, Xuzhou, Jiangsu, China
| | - Yuanhu Yao
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
16
|
Kong Y, Zhao X, Xu M, Pan J, Ma Y, Zou L, Peng Q, Zhang J, Su C, Xu Z, Zhou W, Peng Y, Yang J, Zhou C, Li Y, Guo Q, Chen G, Wu H, Xing P, Zhang L. PD-1 Inhibitor Combined With Radiotherapy and GM-CSF (PRaG) in Patients With Metastatic Solid Tumors: An Open-Label Phase II Study. Front Immunol 2022; 13:952066. [PMID: 35874780 PMCID: PMC9304897 DOI: 10.3389/fimmu.2022.952066] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/10/2022] [Indexed: 01/22/2023] Open
Abstract
Patients with metastatic cancer refractory to standard systemic therapies have a poor prognosis and few therapeutic options. Radiotherapy can shape the tumor microenvironment (TME) by inducing immunogenic cell death and promoting tumor recognition by natural killer cells and T lymphocytes. Granulocyte macrophage-colony stimulating factor (GM-CSF) was known to promote dendric cell maturation and function, and might also induce the macrophage polarization with anti-tumor capabilities. A phase II trial (ChiCTR1900026175) was conducted to assess the clinical efficacy and safety of radiotherapy, PD-1 inhibitor and GM-CSF (PRaG regimen). This trial was registered at http://www.chictr.org.cn/index.aspx. A PRaG cycle consisted of 3 fractions of 5 or 8 Gy delivered for one metastatic lesion from day 1, followed by 200 μg subcutaneous injection of GM-CSF once daily for 2 weeks, and intravenous infusion of PD-1 inhibitor once within one week after completion of radiotherapy. The PRaG regimen was repeated every 21 days for at least two cycles. Once the PRaG therapy was completed, the patient continued PD-1 inhibitor monotherapy until confirmed disease progression or unacceptable toxicity. The primary endpoint was objective response rate (ORR). A total of 54 patients were enrolled with a median follow-up time of 16.4 months. The ORR was 16.7%, and the disease control rate was 46.3% in intent-to-treat patients. Median progression-free survival was 4.0 months (95% confidence interval [CI], 3.3 to 4.8), and median overall survival was 10.5 months (95% CI, 8.7 to 12.2). Grade 3 treatment-related adverse events occurred in five patients (10.0%) and grade 4 in one patient (2.0%). Therefore, the PRaG regimen was well tolerated with acceptable toxicity and may represent a promising salvage treatment for patients with chemotherapy-refractory solid tumors. It is likely that PRaG acts via heating upthe TME with radiotherapy and GM-CSF, which was further boosted by PD-1 inhibitors.
Collapse
Affiliation(s)
- Yuehong Kong
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiangrong Zhao
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Meiling Xu
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Pan
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yifu Ma
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Zou
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiliang Peng
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junjun Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cunjin Su
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhi Xu
- Medical Affairs, ICON Public limited company (ICON Plc), Beijing, China
| | - Wei Zhou
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yong Peng
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiabao Yang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chengliang Zhou
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yujia Li
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiuchen Guo
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Guangqiang Chen
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongya Wu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Key Laboratory for Tumor Immunology of Digestive Tract, Suzhou, China
| | - Pengfei Xing
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Liyuan Zhang, ; Pengfei Xing,
| | - Liyuan Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy & Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Liyuan Zhang, ; Pengfei Xing,
| |
Collapse
|
17
|
Woody S, Hegde A, Arastu H, Peach MS, Sharma N, Walker P, Ju AW. Survival Is Worse in Patients Completing Immunotherapy Prior to SBRT/SRS Compared to Those Receiving It Concurrently or After. Front Oncol 2022; 12:785350. [PMID: 35692764 PMCID: PMC9184512 DOI: 10.3389/fonc.2022.785350] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 04/21/2022] [Indexed: 12/30/2022] Open
Abstract
Purpose/ObjectivesThe abscopal effect could theoretically be potentiated when combined with immunomodulating drugs through increased antigen production. The optimal dosing and schedule of radiotherapy with immunotherapy are unknown, although they are actively investigated in laboratory and clinical models. Clinical data in patients treated for metastatic disease with both modalities may guide future studies.Materials and MethodsThis is a single-institution retrospective review of all patients treated with stereotactic body radiotherapy (SBRT)/stereotactic radiosurgery (SRS) and immunomodulating therapy within 6 months before or after SBRT/SRS for metastatic cancer. Clinical and tumor characteristics were recorded, as well as SBRT/SRS details, immunotherapy details, and survival. Log-rank tests on Kaplan–Meier curves for overall survival (OS) that were calculated from the end of SBRT/SRS were used in univariate analysis and Cox proportional hazards regression for multivariate analysis.ResultsA total of 125 patients were identified who met the inclusion criteria; 70 received SBRT, and 57 received SRS. Eighty-three patients were treated for non-small cell lung cancer, 7 patients for small cell lung cancer, and 35 patients for other cancers, with the most common one being melanoma. Fifty-three percent of patients received nivolumab, 29% pembrolizumab, 13% atezolizumab, 5% other. Twenty percent received immunotherapy before SBRT/SRS, 39% during SBRT/SRS, 41% after. Eighty-six patients had died by the time of the analysis; the median OS for the whole cohort was 9.7 months. Patients who had completed immunotherapy prior to SBRT/SRS had worse OS than those who received concurrent therapy or immunotherapy after SBRT/SRS, with a difference in median OS of 3.6 months vs. 13.0 months (p = 0.010) that was retained on multivariate analysis (p = 0.011). There was no significant difference in OS between patients receiving SRS vs. SBRT (p = 0.20), sex (p = 0.53), age >62 years (p = 0.76), or lung primary vs. others (p = 0.73) on univariate or multivariate analysis. When comparing before/concurrent to after/concurrent administration, there is a difference in survival with after/concurrent survival of 8.181 months and before survival of 13.010 months, but this was not significant (p = 0.25).ConclusionsOS appears to be worse in patients who complete immunotherapy prior to SBRT/SRS compared to those receiving it concurrently or after. The design of this retrospective review may be prone to lead time bias, although the difference in median survival is longer than the 6-month window before SBRT/SRS and could only account for part of this difference. Further analysis into causes of death and toxicity and prospective studies are needed to confirm the results of this analysis.
Collapse
Affiliation(s)
- Susan Woody
- Department of Radiation Oncology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Aparna Hegde
- Department of Radiation Oncology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
- Department of Hematology and Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - Hyder Arastu
- Department of Radiation Oncology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - M. Sean Peach
- Department of Radiation Oncology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Nitika Sharma
- Anne Arundel Medical Center’s DeCesaris Cancer Institute, Annapolis Oncology Center, Annapolis, MD, United States
| | - Paul Walker
- Brody School of Medicine at East Carolina University, Greenville, NC, United States
- Circulogene, Birmingham, AL, United States
| | - Andrew W. Ju
- Department of Radiation Oncology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
- *Correspondence: Andrew W. Ju,
| |
Collapse
|
18
|
Palumbo I, Pasqualetti F, Delishaj D, Gonnelli A, Aristei C, Borghesi S, Pirtoli L, Belgioia L, Arcangeli S. Integrating stereotactic radiotherapy and systemic therapies. Rep Pract Oncol Radiother 2022; 27:310-317. [PMID: 36299395 PMCID: PMC9591045 DOI: 10.5603/rpor.a2022.0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/16/2022] [Indexed: 12/03/2022] Open
Abstract
This paper focuses on stereotactic radiotherapy (SRT ) interactions with targeted therapies and immune system modulating agents because SRT inevitably interacts with them in the treatment of oligometastatic patients. Radiation oncologists need to be aware of the advantages and risks of these interactions which can, on one hand, enhance the effect of therapy or, on the other, potentiate reciprocal toxicities. To date, few prospective studies have evaluated the interactions of SRT with new-generation drugs and data are mainly based on retrospective experiences, which are often related to small sample sizes.
Collapse
Affiliation(s)
- Isabella Palumbo
- Radiation Oncology Section, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - Francesco Pasqualetti
- Department of Radiation Oncology, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Durim Delishaj
- Department of Radiation Oncology, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Alessandra Gonnelli
- Department of Radiation Oncology, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - Simona Borghesi
- Radiation Oncology Unit of Arezzo-Valdarno, Azienda USL Toscana Sud Est, Italy
| | - Luigi Pirtoli
- Unit of Radiation Oncology, University Hospital of Siena, Siena, Italy
| | - Liliana Belgioia
- Radiation Oncology Department, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Stefano Arcangeli
- Department of Radiation Oncology, Policlinico S. Gerardo and University of Milan Bicocca, Milan, Italy
| |
Collapse
|
19
|
Zhou S, Sun X, Jin Z, Yang H, Ye W. The role of autophagy in initiation, progression, TME modification, diagnosis, and treatment of esophageal cancers. Crit Rev Oncol Hematol 2022; 175:103702. [PMID: 35577254 DOI: 10.1016/j.critrevonc.2022.103702] [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: 01/26/2022] [Revised: 04/14/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022] Open
Abstract
Autophagy is a highly conserved metabolic process with a cytoprotective function. Autophagy is involved in cancer, infection, immunity, and inflammation and may be a potential therapeutic target. Increasing evidence has revealed that autophagy has primary implications for esophageal cancer, including its initiation, progression, tumor microenvironment (TME) modification, diagnosis, and treatment. Notably, autophagy displayed excellent application potential in radiotherapy combined with immunotherapy. Radiotherapy combined with immunotherapy is a new potential therapeutic strategy for cancers, including esophageal cancer. Autophagy modulators can work as adjuvant enhancers in radiotherapy or immunotherapy of cancers. This review highlights the most recent data related to the role of autophagy regulation in esophageal cancer.
Collapse
Affiliation(s)
- Suna Zhou
- Department of Radiation Oncology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, P.R. China; Laboratory of Cellular and Molecular Radiation Oncology, The Affiliated Taizhou Hospital, Wenzhou Medical University, Taizhou 317000, Zhejiang, P.R. China; Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou, P.R. China
| | - Xuefeng Sun
- Laboratory of Cellular and Molecular Radiation Oncology, The Affiliated Taizhou Hospital, Wenzhou Medical University, Taizhou 317000, Zhejiang, P.R. China; Department of Radiation Oncology, The Affiliated Taizhou Hospital, Wenzhou Medical University, Taizhou, P.R. China
| | - Zhicheng Jin
- Laboratory of Cellular and Molecular Radiation Oncology, The Affiliated Taizhou Hospital, Wenzhou Medical University, Taizhou 317000, Zhejiang, P.R. China; Department of Radiation Oncology, The Affiliated Taizhou Hospital, Wenzhou Medical University, Taizhou, P.R. China
| | - Haihua Yang
- Laboratory of Cellular and Molecular Radiation Oncology, The Affiliated Taizhou Hospital, Wenzhou Medical University, Taizhou 317000, Zhejiang, P.R. China; Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou, P.R. China; Department of Radiation Oncology, The Affiliated Taizhou Hospital, Wenzhou Medical University, Taizhou, P.R. China
| | - Wenguang Ye
- Department of Gastroenterology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.
| |
Collapse
|
20
|
The Effect of Hyperthermia and Radiotherapy Sequence on Cancer Cell Death and the Immune Phenotype of Breast Cancer Cells. Cancers (Basel) 2022; 14:cancers14092050. [PMID: 35565180 PMCID: PMC9103710 DOI: 10.3390/cancers14092050] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/25/2022] [Accepted: 04/10/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Hyperthermia (HT) is a cancer treatment which locally heats the tumor to supraphysiological temperature, and it is an effective sensitizer for radiotherapy (RT) and chemotherapy. HT is further capable of modulating the immune system. Thus, a better understanding of its effect on the immune phenotype of tumor cells, and particularly when combined with RT, would help to optimize combined anti-cancer treatments. Since in clinics, no standards about the sequence of RT and HT exist, we analyzed whether this differently affects the cell death and immunological phenotype of human breast cancer cells. We revealed that the sequence of HT and RT does not strongly matter from the immunological point of view, however, when HT is combined with RT, it changes the immunophenotype of breast cancer cells and also upregulates immune suppressive immune checkpoint molecules. Thus, the additional application of immune checkpoint inhibitors with RT and HT should be beneficial in clinics. Abstract Hyperthermia (HT) is an accepted treatment for recurrent breast cancer which locally heats the tumor to 39–44 °C, and it is a very potent sensitizer for radiotherapy (RT) and chemotherapy. However, currently little is known about how HT with a distinct temperature, and particularly, how the sequence of HT and RT changes the immune phenotype of breast cancer cells. Therefore, human MDA-MB-231 and MCF-7 breast cancer cells were treated with HT of different temperatures (39, 41 and 44 °C), alone and in combination with RT (2 × 5 Gy) in different sequences, with either RT or HT first, followed by the other. Tumor cell death forms and the expression of immune checkpoint molecules (ICMs) were analyzed by multicolor flow cytometry. Human monocyte-derived dendritic cells (moDCs) were differentiated and co-cultured with the treated cancer cells. In both cell lines, RT was the main stressor for cell death induction, with apoptosis being the prominent cell death form in MCF-7 cells and both apoptosis and necrosis in MDA-MB-231 cells. Here, the sequence of the combined treatments, either RT or HT, did not have a significant impact on the final outcome. The expression of all of the three examined immune suppressive ICMs, namely PD-L1, PD-L2 and HVEM, was significantly increased on MCF-7 cells 120 h after the treatment of RT with HT of any temperature. Of special interest for MDA-MB-231 cells is that only combinations of RT with HT of both 41 and 44 °C induced a significantly increased expression of PD-L2 at all examined time points (24, 48, 72, and 120 h). Generally, high dynamics of ICM expression can be observed after combined RT and HT treatments. There was no significant difference between the different sequences of treatments (either HT + RT or RT + HT) in case of the upregulation of ICMs. Furthermore, the co-culture of moDCs with tumor cells of any treatment had no impact on the expression of activation markers. We conclude that the sequence of HT and RT does not strongly affect the immune phenotype of breast cancer cells. However, when HT is combined with RT, it results in an increased expression of distinct immune suppressive ICMs that should be considered by including immune checkpoint inhibitors in multimodal tumor treatments with RT and HT. Further, combined RT and HT affects the immune system in the effector phase rather than in the priming phase.
Collapse
|
21
|
Swamy K. Stereotactic Body Radiotherapy Immunological Planning-A Review With a Proposed Theoretical Model. Front Oncol 2022; 12:729250. [PMID: 35155221 PMCID: PMC8826062 DOI: 10.3389/fonc.2022.729250] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 01/03/2022] [Indexed: 12/20/2022] Open
Abstract
In the stereotactic body radiotherapy (SBRT) and immunotherapy era, we are moving toward an “immunological radiation plan”, i.e., radiation scheduling with abscopal effect as a vital endpoint as well. The literature review of part A enumerates the advantages of the intermediate dose of SBRT 6–10 Gy per fraction, appropriate use of dose painting, proper timing with immunotherapy, and the potential of immunoadjuvants to maximize cell kill in the irradiated lesions, found to have improved the abscopal effects. Part B summarizes part A, primarily the findings of animal trials, forming the basis of the tenets of the proposed model given in part C to realize the true abscopal potential of the SBRT tumor cell kill of the index lesions. Part C proposes a theoretical model highlighting tumor vasculature integrity as the central theme for converting “abscopal effect by chance” to “abscopal effect by design” using a harmonized combinatorial approach. The proposed model principally deals with the use of SBRT in strategizing increased cell kill in irradiated index tumors along with immunomodulators as a basis for improving the consistency of the abscopal effect. Included is the possible role of integrating immunotherapy just after SBRT, “cyclical” antiangiogenics, and immunoadjuvants/immune metabolites as abscopal effect enhancers of SBRT tumor cell kill. The proposed model suggests convergence research in adopting existing numerous SBRT abscopal enhancing strategies around the central point of sustained vascular integrity to develop decisive clinical trial protocols in the future.
Collapse
|
22
|
Particle radiotherapy and molecular therapies: mechanisms and strategies towards clinical applications. Expert Rev Mol Med 2022; 24:e8. [PMID: 35101155 DOI: 10.1017/erm.2022.2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Immunotherapy and targeted therapy are now commonly used in clinical trials in combination with radiotherapy for several cancers. While results are promising and encouraging, the molecular mechanisms of the interaction between the drugs and radiation remain largely unknown. This is especially important when switching from conventional photon therapy to particle therapy using protons or heavier ions. Different dose deposition patterns and molecular radiobiology can in fact modify the interaction with drugs and their effectiveness. We will show here that whilst the main molecular players are the same after low and high linear energy transfer radiation exposure, significant differences are observed in post-exposure signalling pathways that may lead to different effects of the drugs. We will also emphasise that the problem of the timing between drug administration and radiation and the fractionation regime are critical issues that need to be addressed urgently to achieve optimal results in combined treatments with particle therapy.
Collapse
|
23
|
Scarborough JA, Scott JG. Translation of Precision Medicine Research Into Biomarker-Informed Care in Radiation Oncology. Semin Radiat Oncol 2022; 32:42-53. [PMID: 34861995 PMCID: PMC8667861 DOI: 10.1016/j.semradonc.2021.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The reach of personalized medicine in radiation oncology has expanded greatly over the past few decades as technical precision has improved the delivery of radiation to each patient's unique anatomy. Yet, the consideration of biological heterogeneity between patients has largely not been translated to clinical care. There are innumerable promising advancements in the discovery and validation of biomarkers, which could be used to alter radiation therapy directly or indirectly. Directly, biomarker-informed care may alter treatment dose or identify patients who would benefit most from radiation therapy and who could safely avoid more aggressive care. Indirectly, a variety of biomarkers could assist with choosing the best radiosensitizing chemotherapies. The translation of these advancements into clinical practice will bring radiation oncology even further into the era of precision medicine, treating patients according to their unique anatomical and biological differences.
Collapse
Affiliation(s)
- Jessica A Scarborough
- Translational Hematology and Oncology Research Department, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland,OH; Systems Biology and Bioinformatics Program, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Jacob G Scott
- Translational Hematology and Oncology Research Department, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland,OH; Radiation Oncology Department, Taussig Cancer Institute, Cleveland Clinic Foundation, 10201 Carnegie Ave, Cleveland, OH.
| |
Collapse
|
24
|
Kim H, Lee JH, Kim HJ, Park CM, Wu HG, Goo JM. Extended application of a CT-based artificial intelligence prognostication model in patients with primary lung cancer undergoing stereotactic ablative radiotherapy. Radiother Oncol 2021; 165:166-173. [PMID: 34748856 DOI: 10.1016/j.radonc.2021.10.022] [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: 05/21/2021] [Revised: 09/24/2021] [Accepted: 10/28/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND PURPOSE To validate a computed tomography (CT)-based deep learning prognostication model, originally developed for a surgical cohort, in patients with primary lung cancer undergoing stereotactic ablative radiotherapy (SABR). MATERIALS AND METHODS This retrospective study identified patients with clinical stage T1-2N0M0 lung cancer treated with SABR between 2013 and 2018. The outcomes were local recurrence-free survival (LRFS), disease-free survival (DFS), and overall survival (OS). The discrimination performance of the model, which extracted a quantitative score of cumulative risk for an adverse event up to 900 days, was evaluated using time-dependent receiver operating characteristic curve analysis. Multivariable Cox regression was performed to investigate the independent prognostic value of the model output adjusting for clinical factors including age, sex, smoking status, and clinical T category. RESULTS In total, 135 patients (median age, 78 years; 101 men; 78 [57.8%] adenocarcinomas and 57 [42.2%] squamous cell carcinomas) were evaluated. Most patients (117/135) were treated with 48-60 Gy in four fractions. Median biologically effective dose was 150.0 Gy (interquartile range, 126.9, 150.0 Gy). For LRFS, the area under the curve (AUC) was 0.72 (95% confidence interval [CI]: 0.58, 0.87). The AUCs were 0.70 (95% CI: 0.60, 0.81) for DFS and 0.66 (95% CI: 0.54, 0.77) for OS. Model output was associated with LRFS (adjusted hazard ratio [HR], 1.043; 95% CI: 1.003, 1.085; P = 0.04), DFS (adjusted HR, 1.03; 95% CI: 1.01, 1.05; P = 0.008), and OS (adjusted HR, 1.025; 95% CI: 1.002, 1.047; P = 0.03). CONCLUSION This study showed external validity and transportability of the CT-based deep learning prognostication model for radiotherapy candidates.
Collapse
Affiliation(s)
- Hyungjin Kim
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Hospital, Republic of Korea
| | - Joo Ho Lee
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Hospital, Republic of Korea; Institute of Radiation Medicine, Seoul National University Medical Research Center, Republic of Korea; Cancer Research Institute, Seoul National University, Republic of Korea.
| | - Hak Jae Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Hospital, Republic of Korea; Institute of Radiation Medicine, Seoul National University Medical Research Center, Republic of Korea; Cancer Research Institute, Seoul National University, Republic of Korea
| | - Chang Min Park
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Hospital, Republic of Korea; Institute of Radiation Medicine, Seoul National University Medical Research Center, Republic of Korea; Cancer Research Institute, Seoul National University, Republic of Korea
| | - Hong-Gyun Wu
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Hospital, Republic of Korea; Institute of Radiation Medicine, Seoul National University Medical Research Center, Republic of Korea; Cancer Research Institute, Seoul National University, Republic of Korea
| | - Jin Mo Goo
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Hospital, Republic of Korea; Institute of Radiation Medicine, Seoul National University Medical Research Center, Republic of Korea; Cancer Research Institute, Seoul National University, Republic of Korea
| |
Collapse
|
25
|
Bauman J, Borghaei H. To Give or Not to Give: Consolidative Durvalumab in EGFR-Mutant NSCLC. J Thorac Oncol 2021; 16:894-896. [PMID: 34034885 DOI: 10.1016/j.jtho.2021.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Jessica Bauman
- Department of Hematology-Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Hossein Borghaei
- Department of Hematology-Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| |
Collapse
|
26
|
Allaeys T, Berzenji L, Van Schil PE. Surgery after Induction Targeted Therapy and Immunotherapy for Lung Cancer. Cancers (Basel) 2021; 13:2603. [PMID: 34073302 PMCID: PMC8199385 DOI: 10.3390/cancers13112603] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/25/2022] Open
Abstract
Multimodality therapy for locally advanced non-small cell lung cancer (NSCLC) is a complex and controversial issue, especially regarding optimal treatment regimens for patients with ipsilateral positive mediastinal nodes (N2 disease). Many trials investigating neoadjuvant immunotherapy and targeted therapy in this subpopulation have shown promising results, although concerns have risen regarding surgical feasibility. A thorough literature review was performed, analyzing all recent studies regarding surgical morbidity and mortality. Despite the fact that two major trials investigating this subject were terminated early, the overall consensus is that surgical management seems feasible. However, dissection of hilar vessels may be challenging due to hilar fibrosis. Further research is necessary to identify the role of surgery in these multimodality treatment regimens, and to define matters such as the optimal treatment regimen, the dosage of the different agents used, the interval between induction therapy and surgery, and the role of adjuvant therapy.
Collapse
Affiliation(s)
| | | | - Paul E. Van Schil
- Department of Thoracic and Vascular Surgery, Antwerp University Hospital, Drie Eikenstraat 655, B-2650 Edegem, Antwerp, Belgium; (T.A.); (L.B.)
| |
Collapse
|
27
|
Radiation for Oligometastatic Lung Cancer in the Era of Immunotherapy: What Do We (Need to) Know? Cancers (Basel) 2021; 13:cancers13092132. [PMID: 33925139 PMCID: PMC8125691 DOI: 10.3390/cancers13092132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/21/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary The historical standard treatment of metastatic non-small cell lung cancer (NSCLC) consisted of palliative chemotherapy, with limited influence on survival. With the introduction of immuno- and targeted therapy, the prognosis improved largely. A subset of NSCLC patients with limited metastatic disease, called oligometastatic, might obtain long-term survival by adding a local ablative treatment on all visible disease sites, in addition to the standard systemic treatment. The evidence for this combined treatment is still scarce and comes mainly from the pre-immunotherapy era. As radiotherapy might stimulate the immune system making immunotherapy more efficient, here we review the evidence before and in the era of immunotherapy, and discuss the challenges and prospects of the combined treatment. Abstract Oligometastatic cancer is recognized as a separate entity within the spectrum of metastatic disease. It was suggested that patients with oligometastatic disease can obtain long-term survival by giving local ablative therapy (LAT) to all visible disease locations. However, the true extent from which metastatic cancer should be called “oligometastatic” is unknown, although a consensus definition for oligometastatic disease is proposed by research organizations, such as the EORTC (maximum of five metastases in three organs). Different states of the oligometastatic disease are defined, such as synchronous vs. metachronous, oligopersistent vs. oligoprogressive disease. All clinical trials including patients with non-small cell lung cancer (NSCLC) are small and most are not randomized. Two small randomized phase II trials on synchronous disease showed an improvement in progression free survival, with the addition of LAT, and one also demonstrated an overall survival benefit. Immune checkpoint inhibitors (ICI) were not part of the treatment in these trials, while ICI significantly improved long-term outcomes of patients with metastatic NSCLC. Radiotherapy might improve the prognosis of patients treated with ICI because of its immunostimulatory effects and the possibility to eradicate metastatic deposits. Here, we summarize the data for adding ablative radiotherapy to the treatment of oligometastatic NSCLC, especially in the ICI era, and discuss the challenges of combined treatment.
Collapse
|
28
|
Ohta K, Shimohira M, Ogino H, Nagai K, Sawada Y, Nakayama K, Shibamoto Y. Safety and utility of performing CT-guided biopsies of pulmonary lesions that arise after radiotherapy. J Med Imaging Radiat Oncol 2021; 65:317-322. [PMID: 33733617 DOI: 10.1111/1754-9485.13172] [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: 11/30/2020] [Revised: 01/18/2021] [Accepted: 02/25/2021] [Indexed: 11/29/2022]
Abstract
This study aimed to evaluate the feasibility, safety and usefulness of performing computed tomography (CT)-guided biopsies of pulmonary lesions that arise after radiotherapy. Seventeen patients (14 males and 3 females; median age: 69 years, range: 48-84 years) underwent CT-guided biopsies of pulmonary lesions that occurred in lung regions that had previously been treated with radiotherapy. Three patients underwent CT-guided biopsies twice, and thus, the total number of procedures was 20. We reviewed the subjects' medical records and images, and evaluated the rate for obtaining pathological diagnosis with the biopsy sample, subsequent clinical course, and complications associated with the procedure. In 19 of 20 procedures (95%), the CT-guided biopsy resulted in a pathological diagnosis being obtained. In 14 procedures, the pathological results were consistent with the patients' clinical courses. In the remaining 5 procedures, the lesions were pathologically diagnosed as benign, but they increased in size thereafter; so the lesions were considered to be clinically malignant. The results were considered to represent sampling errors. There were 3 minor complications (slight pneumothorax which did not require drainage) (3/20, 15%), and there were no major complications. In conclusion, performing CT-guided biopsies of pulmonary lesions that arise after radiotherapy appears to be feasible, safe and useful.
Collapse
Affiliation(s)
- Kengo Ohta
- Department of Radiology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Masashi Shimohira
- Department of Radiology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroyuki Ogino
- Department of Radiation Oncology, Nagoya Proton Therapy Center, Nagoya City West Medical Center, Nagoya, Japan
| | - Keiichi Nagai
- Department of Radiology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Yusuke Sawada
- Department of Radiology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Keita Nakayama
- Department of Radiology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuta Shibamoto
- Department of Radiology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
29
|
Usunier B, Brossard C, L’Homme B, Linard C, Benderitter M, Milliat F, Chapel A. HGF and TSG-6 Released by Mesenchymal Stem Cells Attenuate Colon Radiation-Induced Fibrosis. Int J Mol Sci 2021; 22:ijms22041790. [PMID: 33670243 PMCID: PMC7916908 DOI: 10.3390/ijms22041790] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/27/2021] [Accepted: 02/05/2021] [Indexed: 12/18/2022] Open
Abstract
Fibrosis is a leading cause of death in occidental states. The increasing number of patients with fibrosis requires innovative approaches. Despite the proven beneficial effects of mesenchymal stem cell (MSC) therapy on fibrosis, there is little evidence of their anti-fibrotic effects in colorectal fibrosis. The ability of MSCs to reduce radiation-induced colorectal fibrosis has been studied in vivo in Sprague–Dawley rats. After local radiation exposure, rats were injected with MSCs before an initiation of fibrosis. MSCs mediated a downregulation of fibrogenesis by a control of extra cellular matrix (ECM) turnover. For a better understanding of the mechanisms, we used an in vitro model of irradiated cocultured colorectal fibrosis in the presence of human MSCs. Pro-fibrotic cells in the colon are mainly intestinal fibroblasts and smooth muscle cells. Intestinal fibroblasts and smooth muscle cells were irradiated and cocultured in the presence of unirradiated MSCs. MSCs mediated a decrease in profibrotic gene expression and proteins secretion. Silencing hepatocyte growth factor (HGF) and tumor necrosis factor-stimulated gene 6 (TSG-6) in MSCs confirmed the complementary effects of these two genes. HGF and TSG-6 limited the progression of fibrosis by reducing activation of the smooth muscle cells and myofibroblast. To settle in vivo the contribution of HGF and TSG-6 in MSC-antifibrotic effects, rats were treated with MSCs silenced for HGF or TSG-6. HGF and TSG-6 silencing in transplanted MSCs resulted in a significant increase in ECM deposition in colon. These results emphasize the potential of MSCs to influence the pathophysiology of fibrosis-related diseases, which represent a challenging area for innovative treatments.
Collapse
|