1
|
Bylicky MA, Shankavaram U, Aryankalayil MJ, Chopra S, Naz S, Sowers AL, Choudhuri R, Calvert V, Petricoin EF, Eke I, Mitchell JB, Coleman CN. Multiomic-Based Molecular Landscape of FaDu Xenograft Tumors in Mice after a Combinatorial Treatment with Radiation and an HSP90 Inhibitor Identifies Adaptation-Induced Targets of Resistance and Therapeutic Intervention. Mol Cancer Ther 2024; 23:577-588. [PMID: 38359816 PMCID: PMC10985469 DOI: 10.1158/1535-7163.mct-23-0796] [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: 11/15/2023] [Revised: 01/10/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Treatments involving radiation and chemotherapy alone or in combination have improved patient survival and quality of life. However, cancers frequently evade these therapies due to adaptation and tumor evolution. Given the complexity of predicting response based solely on the initial genetic profile of a patient, a predetermined treatment course may miss critical adaptation that can cause resistance or induce new targets for drug and immunotherapy. To address the timescale for these evasive mechanisms, using a mouse xenograft tumor model, we investigated the rapidity of gene expression (mRNA), molecular pathway, and phosphoproteome changes after radiation, an HSP90 inhibitor, or combination. Animals received radiation, drug, or combination treatment for 1 or 2 weeks and were then euthanized along with a time-matched untreated group for comparison. Changes in gene expression occur as early as 1 week after treatment initiation. Apoptosis and cell death pathways were activated in irradiated tumor samples. For the HSP90 inhibitor and combination treatment at weeks 1 and 2 compared with Control Day 1, gene-expression changes induced inhibition of pathways including invasion of cells, vasculogenesis, and viral infection among others. The combination group included both drug-alone and radiation-alone changes. Our data demonstrate the rapidity of gene expression and functional pathway changes in the evolving tumor as it responds to treatment. Discovering these phenotypic adaptations may help elucidate the challenges in using sustained treatment regimens and could also define evolving targets for therapeutic efficacy.
Collapse
Affiliation(s)
- Michelle A. Bylicky
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Uma Shankavaram
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Molykutty J. Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Sunita Chopra
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Sarwat Naz
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Anastasia L. Sowers
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Rajani Choudhuri
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Valerie Calvert
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Emanuel F. Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Iris Eke
- Department of Radiation Oncology, Stanford University Medical School, Stanford, California
| | - James B. Mitchell
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - C. Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
- Radiation Research Program, National Cancer Institute, NIH, Rockville, Maryland
| |
Collapse
|
2
|
Cai Z, Gu X, Xie J, Cheng D, Chen J, Cheng J, Ye J, Lv T. Safety and efficacy of thoracic radiotherapy combined with chemo-immunotherapy in patients with extensive-stage small cell lung cancer: a multicenter retrospective analysis. Transl Lung Cancer Res 2023; 12:1987-2000. [PMID: 38025813 PMCID: PMC10654438 DOI: 10.21037/tlcr-23-294] [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: 05/04/2023] [Accepted: 09/14/2023] [Indexed: 12/01/2023]
Abstract
Background Immunotherapy has greatly increased the survival time of patients with extensive-stage small cell lung cancer (ES-SCLC), and is now a standard first-line treatment for these patients. Increasing evidence suggests a possible synergistic effect between immunotherapy and radiotherapy, yet there is a paucity of evidence regarding the efficacy and safety of thoracic radiotherapy (TRT) combined with chemo-immunotherapy for ES-SCLC. Methods The medical records of 78 consecutive patients with ES-SCLC who received TRT in combination with chemo-immunotherapy at Jinling Hospital and Jiangsu Cancer Hospital from January 2019 to January 2023 were retrospectively reviewed. The median overall survival (mOS) time and median progression-free survival (mPFS) time were used to evaluate efficacy, and the incidence of adverse events (AEs) was used to evaluate safety. Results The median follow-up time was 31.9 months, the objective response rate (ORR) was 59%, and the disease control rate (DCR) was 89.8%. The mOS time was 20.0 months, and the 6-month OS rate was 95%. The mPFS time was 9.2 months, and the 6-month PFS rate was 78%. There were no treatment-related deaths. The incidence of pneumonitis was 23.1%, the incidence of radiation esophagitis was 5.1%, and 2 patients experienced high-grade pneumonitis. Primary liver metastasis was a predictor of poor OS and PFS. Patients who received consolidative TRT after chemo-immunotherapy experienced more benefit than those who received TRT as palliative or salvage treatment for superior vena cava syndrome or disease progression. Conclusions TRT is a feasible treatment for patients who receive chemo-immunotherapy for the management of ES-SCLC in consideration of its considerable efficacy and tolerable safety risk. This treatment is especially useful for patients without primary liver metastasis and who receive consolidative TRT after chemo-immunotherapy. Large-scale prospective studies are needed to confirm the efficacy and safety of this treatment modality.
Collapse
Affiliation(s)
- Zijing Cai
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaoling Gu
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jingyuan Xie
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Di Cheng
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jiayan Chen
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Jing Cheng
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jinjun Ye
- Department of Radiotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Tangfeng Lv
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Nanjing Medical University, Nanjing, China
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
3
|
Coleman CN, Eke I, Makinde AY, Chopra S, Demaria S, Formenti SC, Martello S, Bylicky M, Mitchell JB, Aryankalayil MJ. Radiation-induced Adaptive Response: New Potential for Cancer Treatment. Clin Cancer Res 2020; 26:5781-5790. [PMID: 32554542 PMCID: PMC7669567 DOI: 10.1158/1078-0432.ccr-20-0572] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/24/2020] [Accepted: 06/11/2020] [Indexed: 12/20/2022]
Abstract
Radiotherapy is highly effective due to its ability to physically focus the treatment to target the tumor while sparing normal tissue and its ability to be combined with systemic therapy. This systemic therapy can be utilized before radiotherapy as an adjuvant or induction treatment, during radiotherapy as a radiation "sensitizer," or following radiotherapy as a part of combined modality therapy. As part of a unique concept of using radiation as "focused biology," we investigated how tumors and normal tissues adapt to clinically relevant multifraction (MF) and single-dose (SD) radiation to observe whether the adaptations can induce susceptibility to cell killing by available drugs or by immune enhancement. We identified an adaptation occurring after MF (3 × 2 Gy) that induced cell killing when AKT-mTOR inhibitors were delivered following cessation of radiotherapy. In addition, we identified inducible changes in integrin expression 2 months following cessation of radiotherapy that differ between MF (1 Gy × 10) and SD (10 Gy) that remain targetable compared with preradiotherapy. Adaptation is reflected across different "omics" studies, and thus the range of possible molecular targets is not only broad but also time, dose, and schedule dependent. While much remains to be studied about the radiation adaptive response, radiation should be characterized by its molecular perturbations in addition to physical dose. Consideration of the adaptive effects should result in the design of a tailored radiotherapy treatment plan that accounts for specific molecular changes to be targeted as part of precision multimodality cancer treatment.
Collapse
Affiliation(s)
- C Norman Coleman
- Radiation Oncology Branch and Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| | - Iris Eke
- Radiation Oncology Branch and Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Adeola Y Makinde
- Radiation Oncology Branch and Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Sunita Chopra
- Radiation Oncology Branch and Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Sandra Demaria
- Radiation Oncology and Pathology, Weill Cornell Medicine, New York, New York
| | - Silvia C Formenti
- Radiation Oncology and Pathology, Weill Cornell Medicine, New York, New York
| | - Shannon Martello
- Radiation Oncology Branch and Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Michelle Bylicky
- Radiation Oncology Branch and Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - James B Mitchell
- Radiation Oncology Branch and Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Molykutty J Aryankalayil
- Radiation Oncology Branch and Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
4
|
Fractionation-Dependent Radiosensitization by Molecular Targeting of Nek1. Cells 2020; 9:cells9051235. [PMID: 32429458 PMCID: PMC7291120 DOI: 10.3390/cells9051235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 01/13/2023] Open
Abstract
NIMA (never-in-mitosis gene A)-related kinase 1 (Nek1) is shown to impact on different cellular pathways such as DNA repair, checkpoint activation, and apoptosis. Its role as a molecular target for radiation sensitization of malignant cells, however, remains elusive. Stably transduced doxycycline (Dox)-inducible Nek1 shRNA HeLa cervix and siRNA-transfected HCT-15 colorectal carcinoma cells were irradiated in vitro and 3D clonogenic radiation survival, residual DNA damage, cell cycle distribution, and apoptosis were analyzed. Nek1 knockdown (KD) sensitized both cell lines to ionizing radiation following a single dose irradiation and more pronounced in combination with a 6 h fractionation (3 × 2 Gy) regime. For preclinical analyses we focused on cervical cancer. Nek1 shRNA HeLa cells were grafted into NOD/SCID/IL-2Rγc−/− (NSG) mice and Nek1 KD was induced by Dox-infused drinking water resulting in a significant cytostatic effect if combined with a 6 h fractionation (3 × 2 Gy) regime. In addition, we correlated Nek1 expression in biopsies of patients with cervical cancer with histopathological parameters and clinical follow-up. Our results indicate that elevated levels of Nek1 were associated with an increased rate of local or distant failure, as well as with impaired cancer-specific and overall survival in univariate analyses and for most endpoints in multivariable analyses. Finally, findings from The Cancer Genome Atlas (TCGA) validation cohort confirmed a significant association of high Nek1 expression with a reduced disease-free survival. In conclusion, we consider Nek1 to represent a novel biomarker and potential therapeutic target for drug development in the context of optimized fractionation intervals.
Collapse
|
5
|
Microparticles from tumors exposed to radiation promote immune evasion in part by PD-L1. Oncogene 2019; 39:187-203. [PMID: 31467431 PMCID: PMC6937213 DOI: 10.1038/s41388-019-0971-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 07/26/2019] [Accepted: 08/02/2019] [Indexed: 12/21/2022]
Abstract
Radiotherapy induces immune-related responses in cancer patients by various mechanisms. Here, we investigate the immunomodulatory role of tumor-derived microparticles (TMPs)—extracellular vesicles shed from tumor cells—following radiotherapy. We demonstrate that breast carcinoma cells exposed to radiation shed TMPs containing elevated levels of immune-modulating proteins, one of which is programmed death-ligand 1 (PD-L1). These TMPs inhibit cytotoxic T lymphocyte (CTL) activity both in vitro and in vivo, and thus promote tumor growth. Evidently, adoptive transfer of CTLs pre-cultured with TMPs from irradiated breast carcinoma cells increases tumor growth rates in mice recipients in comparison with control mice receiving CTLs pre-cultured with TMPs from untreated tumor cells. In addition, blocking the PD-1-PD-L1 axis, either genetically or pharmacologically, partially alleviates TMP-mediated inhibition of CTL activity, suggesting that the immunomodulatory effects of TMPs in response to radiotherapy is mediated, in part, by PD-L1. Overall, our findings provide mechanistic insights into the tumor immune surveillance state in response to radiotherapy and suggest a therapeutic synergy between radiotherapy and immune checkpoint inhibitors.
Collapse
|
6
|
Abstract
The radiation stress response can have broad impact. In this Failla Award presentation it is discussed in three components using terms relevant to the current political season as to how the radiation stress response can be applied to the benefit for cancer care and as service to society. Of the people refers to the impact of radiation on cells, tissues and patients. The paradigm our laboratory uses is radiation as a drug, called "focused biology", and physics as "nano-IMRT" because at the nanometer level physics and biology merge. By the people refers to how the general population often reacts to the word "radiation" and how the Radiation Research Society can better enable society to deal with the current realities of radiation in our lives. For the people refers to the potential for radiation oncology and radiation sciences to improve the lives of millions of people globally who are now beyond benefits of cancer treatment and research.
Collapse
Affiliation(s)
- C. Norman Coleman
- Associate Director, Radiation Research Program, Division of Cancer Treatment and Diagnosis; Senior Investigator, Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and Senior Medical Advisor, Office of the Assistant Secretary for Preparedness and Response, Department of Health and Human Services, Washington DC
| |
Collapse
|
7
|
Ahmed MM, Narendra A, Prasanna P, Coleman CN, Krishnan S. Current Insights in Radiation Combination Therapies: Influence of Omics and Novel Targeted Agents in Defining New Concepts in Radiation Biology and Clinical Radiation Oncology. Semin Radiat Oncol 2016; 26:251-3. [DOI: 10.1016/j.semradonc.2016.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|