1
|
Gurau A, Yamauchi S, Ecoff K, Rodgers KP, Eshleman JR, Jr CCT, Huang P, Choi J, Forde PM, Anagnostou V, Brock M, Mei Y. PD-L1 pfeRNAs as blood-based predictors of treatment response of unresectable malignant pleural mesothelioma patients administered Durvalumab with cisplatin and pemetrexed as first-line therapy. Noncoding RNA Res 2025; 12:34-41. [PMID: 40093961 PMCID: PMC11910356 DOI: 10.1016/j.ncrna.2025.02.003] [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: 12/08/2024] [Revised: 01/29/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Background A new therapeutic avenue combining Durvalumab with cisplatin-pemetrexed (Durva-CP) has delivered a promising outcome for previously untreated patients with unresectable malignant pleural mesothelioma (MPM) in clinical trials. However, the limited patient response to Durva-CP needs predictors to select optimal candidates and monitor the developed resistance. Protein functional effector sncRNA (pfeRNA) reveals a fundamental mechanism underlying the regulation of protein activity. The common mechanisms underlying durvalumab, cisplatin, and pemetrexed indicate that PD-L1 pfeRNAs (PDLpfeRNAs) are key molecules that control the treatment response. Methods We specified PDLpfeRNAs by sncRNA deep sequencing, confirmed their binding to PD-L1 by immunoprecipitation and reverse pull-down assays, and demonstrated their roles in controlling the interaction behaviors of PD1/L1 through quality-controlled drug development assays. Following the standards required for the CLIA-compliant LDT, we measured their expression levels in 60 plasma biospecimens from 30 unresectable MPM patients enrolled in the PrE0505 Phase II multicenter study. Using the Cox proportional hazards model and Kaplan-Meier analyses, we described their significance in predicting the treatment response of unresectable MPM patients administered Durva-CP as first-line therapy. Results Two PDLpfeRNAs, PDLpfeRNAa and PDLpfeRNAb, were characterized, confirmed to bind to PD-L1, and identified to control the interaction behaviors of PD-1/L1. Their plasma relative expression levels (REL) demonstrated significant prognostic value for both overall survival (p = 0.0019) and progression-free survival (p = 0.019), and the association remained significant after adjusting for histological subtype (HR 2.59, 95 % CI: 1.00-6.70, p = 0.050) and age (HR 1.03, 95 % CI: 0.98-1.07, p = 0.269). Conclusions Plasma PDLpfeRNAs are predictors of treatment response of unresectable MPM patients treated with Durva-CP as first-line therapy to select optimal candidates and monitor the developed resistance.
Collapse
Affiliation(s)
- Andrei Gurau
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 1650 Orleans Street, Baltimore, MD, 21287, USA
| | - Suguru Yamauchi
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 1650 Orleans Street, Baltimore, MD, 21287, USA
- Department of Esophageal and Gastroenterological Surgery, Faculty of Medicine, Juntendo University, 3-1-3, Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan
| | - Kaitlyn Ecoff
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 1650 Orleans Street, Baltimore, MD, 21287, USA
| | - Kristen P. Rodgers
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 1650 Orleans Street, Baltimore, MD, 21287, USA
| | - James R. Eshleman
- The Pathology Molecular Diagnostics Laboratory, Johns Hopkins School of Medicine, 600 N Wolfe St, Baltimore, MD, 21287, USA
| | - Charles Conover Talbot Jr
- Institute for Basic Biomedical Sciences, Johns Hopkins School of Medicine, 733 North Broadway, Baltimore, MD, 21205, USA
| | - Peng Huang
- Department of Oncology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, 600 North Wolfe Street, Baltimore, MD, 21287, USA
| | - Joshua Choi
- Department of Biology, Krieger School of Arts & Sciences, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
| | - Patrick M. Forde
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 1650 Orleans Street, Baltimore, MD, 21287, USA
| | - Valsamo Anagnostou
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 1650 Orleans Street, Baltimore, MD, 21287, USA
| | - Malcolm Brock
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 1650 Orleans Street, Baltimore, MD, 21287, USA
| | - Yuping Mei
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 1650 Orleans Street, Baltimore, MD, 21287, USA
| |
Collapse
|
2
|
Suleiman H, Emerson A, Wilson PM, Mulligan KA, Ladner RD, LaBonte MJ. Harnessing nucleotide metabolism and immunity in cancer: a tumour microenvironment perspective. FEBS J 2025; 292:2155-2172. [PMID: 39308084 PMCID: PMC12062787 DOI: 10.1111/febs.17278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/06/2024] [Accepted: 09/09/2024] [Indexed: 05/11/2025]
Abstract
The tumour microenvironment (TME) is a dynamic nexus where cancer cell metabolism and the immune system intricately converge, with nucleotide metabolism (NM) playing a pivotal role. This review explores the critical function of NM in cancer cell proliferation and its profound influence on the TME and immune landscape. NM is essential for DNA and RNA synthesis and is markedly upregulated in cancer cells to meet the demands of rapid growth. This metabolic rewiring fuels cancer progression, but also shapes the TME, impacting the function and viability of immune cells. The altered nucleotide milieu in the TME can suppress immune response, aiding cancer cell evasion from immune surveillance. Drug discoveries in the field of NM have revealed different therapeutic strategies, including inhibitors of nucleotide synthesis and drugs targeting salvage pathways, which are discussed thoroughly in this review. Furthermore, the emerging strategy of combining NM-targeted therapies with immunotherapies is emphasised, particularly their effect on sensitising tumours to immune checkpoint inhibitors and enhancing overall treatment efficacy. The Human Genome Project paved the way for personalised medicine, countering the established 'one size fits all' approach to cancer treatment. Advances in understanding the TME and NM have spurred interest in personalised therapeutic strategies. This review highlights the potential of leveraging individual tumour metabolic profiles to guide treatment selection, aiming to optimise efficacy and minimise adverse effects. The strategic importance of targeting NM in cancer therapy and its synergistic potential with immunotherapies offers a path towards more effective and personalised cancer treatments.
Collapse
Affiliation(s)
- Hadil Suleiman
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Alexandra Emerson
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | | | | | - Robert D. Ladner
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
- CV6 Therapeutics (NI) LtdBelfastUK
| | - Melissa J. LaBonte
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| |
Collapse
|
3
|
Zhang B, Song Y, Min Q, Cheng W, Wang J, Fu Y, Yin J. The administration sequences of immune checkpoint inhibitors and chemotherapy cause discrete efficacy when treating non-small cell lung cancer: a retrospective study. Front Immunol 2025; 16:1579420. [PMID: 40356932 PMCID: PMC12066507 DOI: 10.3389/fimmu.2025.1579420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) combined with chemotherapy have become a standard first-line treatment for advanced non-small cell lung cancer (NSCLC). However, the optimal sequence of administrating the two treatments remains controversial. Methods This study included advanced NSCLC patients who received ICIs combined with chemotherapy at Renmin Hospital of Wuhan University and Xiangyang Hospital, Hubei University of Chinese Medicine between 1st September 2020 and 30th September 2024. Patients were categorized into the concurrent, immune-chemo, and chemo-immune groups based on different sequences of treatment administration. The primary endpoints evaluated were survival and treatment efficacy. The secondary endpoint assessed was treatment-related adverse events (TRAEs). Results This two-center, retrospective study included 270 NSCLC patients who received ICIs plus chemotherapy. Survival analysis revealed statistically significant differences across treatment groups. The median overall survival (mOS) durations were 636 days (concurrent group), 615 days (immune-chemo group), and 749 days (chemo-immune group), with a log-rank test demonstrating significant intergroup differences (P = 0.0017). Similarly, median progression-free survival (mPFS) showed distinct patterns at 178 days, 180 days, and 216 days for the respective groups (log-rank P = 0.0134). Additionally, the objective response rates (ORRs) for the three groups were 55.82% (72/129), 58.21% (39/67), and 68.92% (51/74), respectively. The incidence of TRAEs of any grade in the concurrent, the immune-chemo, and the chemo-immune groups was 77.52% (100/129), 65.67% (44/67), and 59.46% (44/74) rates, respectively, which was a significant difference (χ²=7.91, P=0.019). Despite patients experiencing Grade 3 or higher TRAEs had extremely poor prognoses, overall, patients who developed any grade of TRAEs had better survival outcomes, particularly those with skin or endocrine toxicity. Conclusions These findings suggest that the administration sequence of chemotherapy followed by ICIs may yield the greatest clinical benefit, providing a basis for clinical decision-making.
Collapse
Affiliation(s)
- Bicheng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuxiao Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qian Min
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiting Cheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Yang Fu
- Department of Oncology and Hematology, Xiangyang Hospital, Hubei University of Chinese Medicine, Xiangyang, China
| | - Jiaxin Yin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Su YT, Tey SF, Lee CT, Lin CY, Tsai JS, Lin CC, Kuo CW. Comparative Survival Analysis of Anti-Angiogenic Agent Plus Immunochemotherapy in NSCLC Patients After Frontline EGFR-TKI Treatment: A Retrospective Cohort Study. Kaohsiung J Med Sci 2025:e70023. [PMID: 40289745 DOI: 10.1002/kjm2.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/14/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Advanced-stage EGFR-mutated lung non-small cell lung cancer (NSCLC) challenges current treatment paradigms, particularly after frontline EGFR-TKI therapy failure. This study investigates the survival impact of combined anti-angiogenic agent and immunochemotherapy (AICT) for this population. We retrospectively analyzed NSCLC patients at National Cheng Kung University Hospital from January 2010 to December 2022, focusing on those who had disease progression beyond frontline EGFR-TKI treatments. Survival outcomes were assessed through progression-free survival (PFS) and overall survival post-TKI failure (OSpTKI). Propensity score was employed to match patients, with Kaplan-Meier curve and multivariable Cox regression analysis determining the survival benefits. Analyses were also performed for subgroups based on PD-L1 level, treatment lines, and regimens. A total of 412 patients were enrolled, with 27 receiving AICT. Compared to patients who did not receive AICT, those who received AICT had longer PFS (5.9 vs. 3.9 months, p = 0.024) and longer OSpTKI (17.9 vs. 11.9 months, p = 0.018). The observed survival advantage in PFS and OSpTKI was consistent in both the original cohort (for PFS: hazard ratio [HR] = 0.59, 95% confidence interval [CI] = 0.39-0.90, p = 0.014; for OSpTKI: HR = 0.41, 95% CI = 0.24-0.69, p < 0.001) and after propensity score matching (for PFS: HR = 0.56, 95% CI = 0.35-0.98, p = 0.014; for OSpTKI: HR = 0.45, 95% CI = 0.26-0.79, p = 0.006). In the subgroup analyses, patients with PD-L1 ≥ 1%, those who received AICT as a second-line therapy, or those treated in conjunction with pemetrexed showed a PFS benefit. AICT improves survival outcomes in advanced-stage EGFR-mutated NSCLC, advocating for its integration into treatment regimens.
Collapse
Affiliation(s)
- Yi-Tse Su
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Farn Tey
- Division of Pulmonary Medicine, Chi-Mei Medical Center, Tainan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chung-Ta Lee
- Department of Pathology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Yu Lin
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jeng-Shiuan Tsai
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Chung Lin
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chin-Wei Kuo
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
5
|
Kicken MP, Deenen MJ, van der Wekken AJ, van den Borne BEEM, van den Heuvel MM, Ter Heine R. Opportunities for Precision Dosing of Cytotoxic Drugs in Non-Small Cell Lung Cancer: Bridging the Gap in Precision Medicine. Clin Pharmacokinet 2025; 64:511-531. [PMID: 40045151 PMCID: PMC12041064 DOI: 10.1007/s40262-025-01492-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2025] [Indexed: 04/30/2025]
Abstract
Precision dosing of classical cytotoxic drugs in oncology remains underdeveloped, especially in treating non-small cell lung cancer (NSCLC). Despite advancements in targeted therapy and immunotherapy, classical cytotoxic agents continue to play a critical role in NSCLC treatment. However, the current body surface area (BSA)-based dosing of these agents fails to adequately address interindividual variability in pharmacokinetics. By better considering patient characteristics, treatment outcomes can be improved, reducing risks of under-exposure and over-exposure. This narrative review explores opportunities for precision dosing for key cytotoxic agents used in NSCLC treatment: cisplatin, carboplatin, pemetrexed, docetaxel, (nab-)paclitaxel, gemcitabine, and vinorelbine. A comprehensive review of regulatory reports and an extensive literature search were conducted to evaluate current dosing practices, pharmacokinetics, pharmacodynamics, and exposure-response relationships. Our findings highlight promising developments in precision dosing, although the number of directly implementable strategies remains limited. The most compelling evidence supports using the biomarker cystatin C for more precise carboplatin dosing and adopting weekly dosing schedules for docetaxel, paclitaxel, and nab-paclitaxel. Additionally, we recommend direct implementation of therapeutic drug monitoring (TDM)-guided dosing for paclitaxel. This review stresses the urgent need to reassess conventional dosing paradigms for classical cytotoxic agents to better align with the principles of the precision dosing framework. Our recommendations show the potential of precision dosing to improve NSCLC treatment, addressing gaps in the current dosing of classical cytotoxic drugs. Given the large NSCLC patient population, optimising the dosing of these agents could significantly improve treatment outcomes and reduce toxicity for many patients.
Collapse
Affiliation(s)
- M P Kicken
- Department of Clinical Pharmacy, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands.
- Department of Pharmacy, Radboudumc, Research Institute for Medical Innovation, Nijmegen, The Netherlands.
| | - M J Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - A J van der Wekken
- Department of Pulmonology and Tuberculosis, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - M M van den Heuvel
- Department of Pulmonology, Radboudumc, Research Institute for Medical Innovation, Nijmegen, The Netherlands
- Department of Pulmonology, University Medical Center, Utrecht, The Netherlands
| | - R Ter Heine
- Department of Pharmacy, Radboudumc, Research Institute for Medical Innovation, Nijmegen, The Netherlands
| |
Collapse
|
6
|
Li X, Xu H, Hong R, Yang H, Xu L, Zheng G, Xie B. Frontline pemetrexed and cisplatin based-chemotherapy combined with NRT promoted the antitumor in a mouse model of lung carcinoma. Int Immunopharmacol 2025; 149:114174. [PMID: 39929101 DOI: 10.1016/j.intimp.2025.114174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025]
Abstract
The efficacy of neoantigen-reactive T cells (NRT) therapy in solid tumors, encompassing aspects such as infiltration, recognition, cytotoxicity, and enduring persistence, is notably influenced by the immunological microenvironment. This study endeavors to investigate whether the co-administration of pemetrexed and cisplatin augments the therapeutic efficacy of NRT therapy in lung cancer. Neoantigens were predicted using a comprehensive analysis of mutation data from Lewis lung carcinoma cells and mouse tail tissues. The immunogenicity of NRT cells was assessed through flow cytometry and IFN-γ ELISpot assays. A mouse model of NSCLC was used to investigate the anti-tumor effects of NRT combined with chemotherapy. The combination of NRT cells and chemotherapy significantly inhibited tumor growth in a mouse model, increased CD3+/CD137+ T cells to promote IFN-γ secretion from NRT cells, and up-regulated the levels of inflammatory cytokine proteins including IFN-γ, TNF, IL-6 and IL-10. Immunofluorescence analysis confirmed increased T-cell infiltration in tumor tissues without adverse effects on vital organs. In addition, transcriptome analyses indicated that the tumor microenvironment was altered to favor M1-like macrophages with an increased M1/M2 ratio, creating a pro-inflammatory environment. The integration of NRT with frontline chemotherapy for lung cancer could yield profoundly ideal therapeutic outcomes.
Collapse
Affiliation(s)
- Xiaoqin Li
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Hang Xu
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Rujun Hong
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Haitao Yang
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Lihuan Xu
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Guanying Zheng
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China.
| | - Baosong Xie
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China.
| |
Collapse
|
7
|
Hsin IL, Chiu LY, Ko JL, Wang PH, Wu PJ. Targeted inhibition of integrin αVβ3 induces cytotoxicity and suppresses migration ability in ovarian cancer cells and tumor spheroids. Int J Med Sci 2025; 22:1544-1554. [PMID: 40093794 PMCID: PMC11905266 DOI: 10.7150/ijms.103141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/09/2025] [Indexed: 03/19/2025] Open
Abstract
Ovarian cancer is a gynecological malignancy that has poor prognosis and high lethality. Integrin αVβ3 is highly expressed in solid cancer cells, including ovarian cancer, and is important in proliferation and cell migration. In this study, we performed two-dimensional (2D) and three‑dimensional (3D) cell culture systems to investigate the potential of integrin αVβ3 as a therapeutic target for ovarian cancer. Inhibition of integrin αVβ3 by antagonist cilengitide (CGT) and shRNA significantly reduce the cell viability of ovarian cancer cells. Co-treatment of CGT and cisplatin induced synergistic cytotoxicity in SKOV3 cells. CGT reduced the protein expressions of phospho-FAK, CD44, and PD-L1. CGT reduced mitochondrial membrane potential and induced apoptotic cell death. To mimic the tumor growth in the extracellular matrix, a tumor spheroid formation assay was performed with Matrigel and epidermal growth factor (EGF). CGT reduced the size of spheroids that grew in 50% Matrigel with or without EGF induction. CGT also enhanced the inhibiting effect of T cells on tumor spheroids. The cell migration ability of SKOV3 cells was blunted by CGT by tumor spheroid-based migration assay. This study used 2D and 3D cell models to provide novel insight into ovarian cancer therapy by targeting integrin αVβ3 and suitable cell models for searching integrin αVβ3-targeting drugs.
Collapse
Affiliation(s)
- I-Lun Hsin
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Nursing, National Taichung University of Science and Technology, Taichung, 40640, Taiwan
- Institute and Department of Food Science, Central Taiwan University of Science and Technology, Taichung, 40601, Taiwan
| | - Ling-Yen Chiu
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Division of Medical Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan
| | - Po-Hui Wang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan
| | - Pei-Ju Wu
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan
| |
Collapse
|
8
|
Liang H, Liang W, Tian J, Zheng S, Su Y, Huang P, Chen R, Guan Z, Cai Q. Safety and efficacy of neoadjuvant therapy with tislelizumab plus chemotherapy for locally advanced head and neck squamous cell carcinoma: a single-arm, retrospective study. Cancer Immunol Immunother 2025; 74:108. [PMID: 39932534 PMCID: PMC11813832 DOI: 10.1007/s00262-025-03953-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a very common type of head and neck cancer, and the 5-year overall survival (OS) rate is only 50%. Inhibitors targeting the programmed death 1 (PD-1) pathway have gained widespread clinical adoption. However, the relationship between the infiltration characteristics of immune cells in the tumor immune microenvironment (TIME) and the effect of immunotherapy on HNSCC remains to be explored. METHODS Patients diagnosed with HNSCC who received Tislelizumab combined with chemotherapy were reviewed between February 2021 and March 30, 2024, in our single center. The laryngoscopy, magnetic resonance imaging (MRI), and pathologic response were evaluated to the efficacy of neoadjuvant therapy with Tislelizumab plus chemotherapy treatment. Treatment-related adverse events (TRAEs) were evaluated according to the Common Terminology Criteria for Adverse Events version 5.0. RESULT Our analysis involved 42 patients who received Tislelizumab combined with chemotherapy. A total of 18 patients underwent surgical treatment following the completion of immunotherapy combined with chemotherapy. Among them, 6 patients achieved pCR (33%). The 1-year OS rate of the 42 patients enrolled in the study was 95.05%, and the 1-year PFS rate was 89.86%. There was a significant positive correlation between the lymphocyte density in HNSCC prior to the administration of neoadjuvant PD-1 inhibitor therapy combined with chemotherapy and the immunotherapy efficacy. Compared with pretreatment, the neutrophil-to-lymphocyte ratio (NLR) was significantly decreased, and the lymphocyte density was significantly increased in HNSCC patients after immunotherapy. CONCLUSIONS The integration of neoadjuvant PD-1 inhibitor therapy with chemotherapy has been demonstrated to be a safe and effective strategy, can improve the tumor response rate and survival rate, and is a valuable treatment for patients with HNSCC. Furthermore, the study suggests that an elevated NLR within the HNSCC tumor microenvironment could potentially serve as a biomarker indicative of diminished immunotherapy efficacy.
Collapse
Affiliation(s)
- Haifeng Liang
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Wenting Liang
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Jiawang Tian
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Shibei Zheng
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
- Department of Otorhinolaryngology, Head and Neck Surgery, Shenzhen Baoan District People's Hospital, Shenzhen, China
| | - Yangzhou Su
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Piao Huang
- Department of Pathology, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Renhui Chen
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China.
| | - Zhong Guan
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China.
| | - Qian Cai
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China.
- Department of Otolaryngology-Head and Neck, Zhujiang Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Qiu Y, Xie E, Xu H, Cheng H, Li G. One-carbon metabolism shapes T cell immunity in cancer. Trends Endocrinol Metab 2024; 35:967-980. [PMID: 38925992 DOI: 10.1016/j.tem.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
One-carbon metabolism (1CM), comprising folate metabolism and methionine metabolism, serves as an important mechanism for cellular energy provision and the production of vital signaling molecules, including single-carbon moieties. Its regulation is instrumental in sustaining the proliferation of cancer cells and facilitating metastasis; in addition, recent research has shed light on its impact on the efficacy of T cell-mediated immunotherapy. In this review, we consolidate current insights into how 1CM affects T cell activation, differentiation, and functionality. Furthermore, we delve into the strategies for modulating 1CM in both T cells and tumor cells to enhance the efficacy of adoptively transferred T cells, overcome metabolic challenges in the tumor microenvironment (TME), and maximize the benefits of T cell-mediated immunotherapy.
Collapse
Affiliation(s)
- Yajing Qiu
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Ermei Xie
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Haipeng Xu
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fujian, 350011, China
| | - Hongcheng Cheng
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| | - Guideng Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
10
|
Wang J, Guo B, Sun Z, Zhao S, Cao L, Zhong Z, Meng F. Polymersomal Poly(I:C) Self-Magnifies Antitumor Immunity by Inducing Immunogenic Cell Death and Systemic Immune Activation. Adv Healthc Mater 2024; 13:e2400784. [PMID: 38896790 DOI: 10.1002/adhm.202400784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/18/2024] [Indexed: 06/21/2024]
Abstract
Immunotherapy has emerged as a powerful weapon against lung cancer, yet only a fraction of patients respond to the treatment. Poly(I:C) (PIC) effectively triggers both innate and adaptive immunity. It can also induce immunogenic cell death (ICD) in tumor cells. However, its efficacy is hindered by its instability in vivo and limited cellular uptake. To address this, PIC is encapsulated in cRGD-functionalized polymersomes (t-PPIC), which significantly increases its stability and uptake, thus activating dendritic cells (DCs) and inducing apoptosis of lung tumor cells in vitro. In a murine LLC lung tumor model, systemic administration of t-PPIC effectively suppresses tumor growth and leads to survival benefits, with 40% of the mice becoming tumor-free. Notably, t-PPIC provokes stronger apoptosis and ICD in tumor tissue and elicits a more potent stimulation of DCs, recruitment of natural killer (NK) cells, and activation of CD8+ T cells, compared to free PIC and nontargeted PPIC controls. Furthermore, when combined with immune checkpoint inhibitors or radiotherapy, t-PPIC amplifies the antitumor immune response, resulting in complete regression in 60% of the mice. These compelling findings underscore the potential of integrin-targeted polymersomal PIC to enhance antitumor immunity by simultaneously inducing ICD and systemic immune activation.
Collapse
Affiliation(s)
- Jingyi Wang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
| | - Beibei Guo
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
| | - Zhiwei Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
| | - Songsong Zhao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
| | - Li Cao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215006, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215006, P. R. China
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, 215006, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215006, P. R. China
| |
Collapse
|
11
|
Nishioka N, Kawachi H, Yamada T, Tamiya M, Negi Y, Goto Y, Nakao A, Shiotsu S, Tanimura K, Takeda T, Okada A, Harada T, Date K, Chihara Y, Hasegawa I, Tamiya N, Masui T, Sai N, Ishida M, Katayama Y, Morimoto K, Iwasaku M, Tokuda S, Kijima T, Takayama K. Unraveling the influence of TTF-1 expression on immunotherapy outcomes in PD-L1-high non-squamous NSCLC: a retrospective multicenter study. Front Immunol 2024; 15:1399889. [PMID: 39076994 PMCID: PMC11284020 DOI: 10.3389/fimmu.2024.1399889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
Introduction Several studies explored the association between thyroid transcription factor-1 (TTF-1) and the therapeutic efficacy of immunotherapy. However, the effect of TTF-1 on the therapeutic efficacy of programmed death-1 (PD-1) inhibitor/chemoimmunotherapy in patients with non-squamous non-small cell lung cancer (non-Sq NSCLC) with a programmed death-ligand 1 (PD-L1) tumor proportion score of 50% or more who are highly susceptible to immunotherapy remains unresolved. Therefore, we evaluated whether TTF-1 has a clinical impact on this population. Methods Patients with non-Sq NSCLC and high PD-L1 expression who received PD-1 inhibitor monotherapy or chemoimmunotherapy between May 2017 and December 2020 were retrospectively enrolled. Treatment efficacy was compared after adjusting for baseline differences using propensity score matching. Results Among the 446 patients with NSCLC with high PD-L1 expression, 266 patients with non-Sq NSCLC were analyzed. No significant differences in therapeutic efficacy were observed between the TTF-1-positive and -negative groups in the overall and propensity score-matched populations. Of chemoimmunotherapy, pemetrexed containing regimen significantly prolonged progression-free survival compared to chemoimmunotherapy without pemetrexed, regardless of TTF-1 expression (TTF1 positive; HR: 0.46 (95% Confidence interval: 0.26-0.81), p<0.01, TTF-1 negative; HR: 0.29 (95% Confidence interval: 0.09-0.93), p=0.02). Discussion TTF-1 expression did not affect the efficacy of PD-1 inhibitor monotherapy or chemoimmunotherapy in patients with non-Sq NSCLC with high PD-L1 expression. In this population, pemetrexed-containing chemoimmunotherapy demonstrated superior anti-tumor efficacy, irrespective of TTF-1 expression.
Collapse
Affiliation(s)
- Naoya Nishioka
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Hayato Kawachi
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Tadaaki Yamada
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Motohiro Tamiya
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Osaka, Japan
| | - Yoshiki Negi
- Department of Respiratory Medicine and Hematology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Yasuhiro Goto
- Department of Respiratory Medicine, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Akira Nakao
- Department of Respiratory Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Shinsuke Shiotsu
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Kyoto, Japan
| | - Keiko Tanimura
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Kyoto, Japan
| | - Takayuki Takeda
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Kyoto, Japan
| | - Asuka Okada
- Department of Respiratory Medicine, Saiseikai Suita Hospital, Suita, Osaka, Japan
| | - Taishi Harada
- Department of Medical Oncology, Fukuchiyama City Hospital, Fukuchiyama, Kyoto, Japan
| | - Koji Date
- Department of Pulmonary Medicine, Kyoto Chubu Medical Center, Nantan, Kyoto, Japan
| | - Yusuke Chihara
- Department of Respiratory Medicine, Uji-Tokushukai Medical Center, Uji, Kyoto, Japan
| | - Isao Hasegawa
- Department of Respiratory Medicine, Saiseikai Shigaken Hospital, Ritto, Shiga, Japan
| | - Nobuyo Tamiya
- Department of Respiratory Medicine, Rakuwakai Otowa Hospital, Kyoto, Kyoto, Japan
| | - Taiki Masui
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Natsuki Sai
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Masaki Ishida
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Yuki Katayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Kenji Morimoto
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Masahiro Iwasaku
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Shinsaku Tokuda
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Takashi Kijima
- Department of Respiratory Medicine and Hematology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| |
Collapse
|
12
|
Mariniello A, Nasti TH, Chang DY, Hashimoto M, Malik S, McManus DT, Lee J, McGuire DJ, Cardenas MA, Umana P, Nicolini V, Antia R, Saha A, Buchwald Z, Kissick H, Ghorani E, Novello S, Sangiolo D, Scagliotti GV, Ramalingam SS, Ahmed R. Platinum-Based Chemotherapy Attenuates the Effector Response of CD8 T Cells to Concomitant PD-1 Blockade. Clin Cancer Res 2024; 30:1833-1845. [PMID: 37992307 PMCID: PMC11061601 DOI: 10.1158/1078-0432.ccr-23-1316] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/27/2023] [Accepted: 11/20/2023] [Indexed: 11/24/2023]
Abstract
PURPOSE Combination of chemotherapy with programmed cell death 1 (PD-1) blockade is a front-line treatment for lung cancer. However, it remains unknown whether and how chemotherapy affects the response of exhausted CD8 T cells to PD-1 blockade. EXPERIMENTAL DESIGN We used the well-established mouse model of T-cell exhaustion with chronic lymphocytic choriomeningitis virus (LCMV) infection to assess the effect of chemotherapy (cisplatin+pemetrexed) on T-cell response to PD-1 blockade, in the absence of the impact of chemotherapy on antigen release and presentation observed in tumor models. RESULTS When concomitantly administered with PD-1 blockade, chemotherapy affected the differentiation path of LCMV-specific CD8 T cells from stem-like to transitory effector cells, thereby reducing their expansion and production of IFNγ. After combination treatment, these restrained effector responses resulted in impaired viral control, compared with PD-1 blockade alone. The sequential combination strategy, where PD-1 blockade followed chemotherapy, proved to be superior to the concomitant combination, preserving the proliferative response of exhausted CD8 T cells to PD-1 blockade. Our findings suggest that the stem-like CD8 T cells themselves are relatively unaffected by chemotherapy partly because they are quiescent and maintained by slow self-renewal at the steady state. However, upon the proliferative burst mediated by PD-1 blockade, the accelerated differentiation and self-renewal of stem-like cells may be curbed by concomitant chemotherapy, ultimately resulting in impaired overall CD8 T-cell effector functions. CONCLUSIONS In a translational context, we provide a proof-of-concept to consider optimizing the timing of chemo-immunotherapy strategies for improved CD8 T-cell functions. See related commentary by Vignali and Luke, p. 1705.
Collapse
Affiliation(s)
- Annapaola Mariniello
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Department of Oncology, University of Torino, Turin, Italy
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Tahseen H. Nasti
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Daniel Y. Chang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Masao Hashimoto
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Sakshi Malik
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Daniel T. McManus
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Judong Lee
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Donald J. McGuire
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Maria A. Cardenas
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia
| | - Pablo Umana
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Valeria Nicolini
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, Georgia
| | - Ananya Saha
- Department of Biology, Emory University, Atlanta, Georgia
| | - Zachary Buchwald
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia
| | - Hayden Kissick
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia
| | - Ehsan Ghorani
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Cancer Immunology and Immunotherapy Unit, Imperial College London, Department of Surgery and Cancer, London, United Kingdom
| | - Silvia Novello
- Department of Oncology, University of Torino, Turin, Italy
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Turin, Italy
| | | | - Suresh S. Ramalingam
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Rafi Ahmed
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
| |
Collapse
|
13
|
Kozai H, Ogino H, Mitsuhashi A, Nguyen NT, Tsukazaki Y, Yabuki Y, Ozaki R, Yoneda H, Sato S, Hanibuchi M, Shinohara T, Nokihara H, Nishioka Y. Potential of fluoropyrimidine to be an immunologically optimal partner of immune checkpoint inhibitors through inducing immunogenic cell death for thoracic malignancies. Thorac Cancer 2024; 15:369-378. [PMID: 38146645 PMCID: PMC10864125 DOI: 10.1111/1759-7714.15200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are a revolutionary paradigm in the treatment of thoracic malignancies and chemoimmunotherapy is a current standard care in this field. Chemotherapeutic agents are known to induce not only direct cytotoxic effects on tumor cells but also immune modulating effects, such as stimulating immunogenic cell death (ICD). Currently, either pemetrexed (PEM) or taxane plus platinum are combined with ICIs for patients with non-small cell lung cancer (NSCLC); however, it is still unknown whether these agents are immunologically optimal partners for ICIs. METHODS To determine the immunologically optimal chemotherapeutic agent, we first evaluated the ability of several chemotherapeutic agents, including platinum, PEM, taxane, and 5-fluorouracil (5-FU) to induce ICD using several thoracic tumor cell lines in vitro. ICD was evaluated by the cell surface expression of calreticulin (CRT) and adenosine-triphosphate (ATP) secretion. We further performed an antitumor vaccination assay in vivo. RESULTS 5-FU induced cell surface expression of CRT and ATP secretion most efficiently among the several chemotherapeutic agents. This effect was enhanced when it was combined with platinum. In the antitumor vaccination assay in vivo, we found that vaccination with dying-AB1-HA (a murine malignant mesothelioma cell line) cells treated with 5-FU, but neither PEM nor PTX, reduced the tumor growth of living-AB1-HA cells inoculated 1 week after vaccination by recruiting CD3+ CD8+ T cells into the tumor microenvironment. CONCLUSION Our findings indicate that fluoropyrimidine can be an immunologically optimal partner of ICIs through the induction of ICD for thoracic malignancies.
Collapse
Affiliation(s)
- Hiroyuki Kozai
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Hirokazu Ogino
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Atsushi Mitsuhashi
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Na Thi Nguyen
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Yuki Tsukazaki
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Yohei Yabuki
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Ryohiko Ozaki
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Hiroto Yoneda
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Seidai Sato
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Masaki Hanibuchi
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
- Department of Community Medicine for RespirologyHematology, and Metabolism, Graduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Tsutomu Shinohara
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
- Department of Community Medicine for RespirologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Hiroshi Nokihara
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
- Respiratory Medicine, Center Hospital of the National Center for Global Health and MedicineTokyoJapan
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| |
Collapse
|
14
|
Ren X, Wang X, Zheng G, Wang S, Wang Q, Yuan M, Xu T, Xu J, Huang P, Ge M. Targeting one-carbon metabolism for cancer immunotherapy. Clin Transl Med 2024; 14:e1521. [PMID: 38279895 PMCID: PMC10819114 DOI: 10.1002/ctm2.1521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND One-carbon (1C) metabolism is a metabolic network that plays essential roles in biological reactions. In 1C metabolism, a series of nutrients are used to fuel metabolic pathways, including nucleotide metabolism, amino acid metabolism, cellular redox defence and epigenetic maintenance. At present, 1C metabolism is considered the hallmark of cancer. The 1C units obtained from the metabolic pathways increase the proliferation rate of cancer cells. In addition, anticancer drugs, such as methotrexate, which target 1C metabolism, have long been used in the clinic. In terms of immunotherapy, 1C metabolism has been used to explore biomarkers connected with immunotherapy response and immune-related adverse events in patients. METHODS We collected numerous literatures to explain the roles of one-carbon metabolism in cancer immunotherapy. RESULTS In this review, we focus on the important pathways in 1C metabolism and the function of 1C metabolism enzymes in cancer immunotherapy. Then, we summarise the inhibitors acting on 1C metabolism and their potential application on cancer immunotherapy. Finally, we provide a viewpoint and conclusion regarding the opportunities and challenges of targeting 1C metabolism for cancer immunotherapy in clinical practicability in the future. CONCLUSION Targeting one-carbon metabolism is useful for cancer immunotherapy.
Collapse
Affiliation(s)
- Xinxin Ren
- Department of Head and Neck SurgeryOtolaryngology & Head and Neck Center, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Malignant TumorHangzhouZhejiangChina
- Department of PathologyCancer CenterZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Xiang Wang
- Department of PharmacyAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Guowan Zheng
- Department of Head and Neck SurgeryOtolaryngology & Head and Neck Center, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Malignant TumorHangzhouZhejiangChina
| | - Shanshan Wang
- Department of PharmacyCenter for Clinical PharmacyCancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Qiyue Wang
- Department of Head and Neck SurgeryOtolaryngology & Head and Neck Center, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Malignant TumorHangzhouZhejiangChina
| | - Mengnan Yuan
- Department of PharmacyCenter for Clinical PharmacyCancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Tong Xu
- Department of PharmacyCenter for Clinical PharmacyCancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Jiajie Xu
- Department of Head and Neck SurgeryOtolaryngology & Head and Neck Center, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Malignant TumorHangzhouZhejiangChina
| | - Ping Huang
- Department of PharmacyCenter for Clinical PharmacyCancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Minghua Ge
- Department of Head and Neck SurgeryOtolaryngology & Head and Neck Center, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Malignant TumorHangzhouZhejiangChina
| |
Collapse
|
15
|
O’Brien Gore C, Billman A, Hunjan S, Colebrook J, Choy D, Li W, Haynes J, Wade J, Hobern E, McDonald L, Papa S, Brugman M, Kordasti S, Montiel-Equihua C. Pre-treatment with systemic agents for advanced NSCLC elicits changes in the phenotype of autologous T cell therapy products. Mol Ther Oncolytics 2023; 31:100749. [PMID: 38075248 PMCID: PMC10701366 DOI: 10.1016/j.omto.2023.100749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 11/07/2023] [Indexed: 04/17/2025] Open
Abstract
The antitumor activity of adoptive T cell therapies (ACT) is highly dependent on the expansion, persistence, and continued activity of adoptively transferred cells. Clinical studies using ACTs have revealed that products that possess and maintain less differentiated phenotypes, including memory and precursor T cells, show increased antitumor efficacy and superior patient outcomes owing to their increased expansion, persistence, and ability to differentiate into effector progeny that elicit antitumor responses. Strategies that drive the differentiation into memory or precursor-type T cell subsets with high potential for persistence and self-renewal will enhance adoptively transferred T cell maintenance and promote durable antitumor efficacy. Because of the high costs associated with ACT manufacturing, ACTs are often only offered to patients after multiple rounds of systemic therapy. An essential factor to consider in producing autologous ACT medicinal products is the impact of the patient's initial T cell fitness and subtype composition, which will likely differ with age, disease history, and treatment with prior anti-cancer therapies. This study evaluated the impact of systemic anti-cancer therapy for non-small cell lung cancer treatment on the T cell phenotype of the patient at baseline and the quality and characteristics of the genetically modified autologous T cell therapy product after manufacturing.
Collapse
Affiliation(s)
- Charlotte O’Brien Gore
- School of Cancer and Pharmaceutical Sciences, King’s College London, Faculty of Life Sciences and Medicine, Guy’s Hospital, London, UK
| | - Amy Billman
- Analytical Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, SG1 2NY Hertfordshire, UK
| | - Suchete Hunjan
- Oncology Cell Therapy Unit, GSK Medicine Research Centre, Stevenage, SG1 2NY Hertfordshire, UK
| | - Jayne Colebrook
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, SG1 2NY Hertfordshire, UK
| | - Desmond Choy
- School of Cancer and Pharmaceutical Sciences, King’s College London, Faculty of Life Sciences and Medicine, Guy’s Hospital, London, UK
| | - Wilson Li
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, SG1 2NY Hertfordshire, UK
| | - Jack Haynes
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, SG1 2NY Hertfordshire, UK
| | - Jennifer Wade
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, SG1 2NY Hertfordshire, UK
| | - Emily Hobern
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, SG1 2NY Hertfordshire, UK
| | - Louisa McDonald
- Oncology & Hematology Clinical Trials (OHCT), Guy’s Hospital, 1st Floor Chapel Wing, Great Maze Pond, SE1 9RT London, UK
| | - Sophie Papa
- School of Cancer and Pharmaceutical Sciences, King’s College London, Faculty of Life Sciences and Medicine, Guy’s Hospital, London, UK
| | - Martijn Brugman
- Analytical Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, SG1 2NY Hertfordshire, UK
| | - Shahram Kordasti
- School of Cancer and Pharmaceutical Sciences, King’s College London, Faculty of Life Sciences and Medicine, Guy’s Hospital, London, UK
| | - Claudia Montiel-Equihua
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, SG1 2NY Hertfordshire, UK
| |
Collapse
|
16
|
Chen Y, Xu J, Liu X, Guo L, Yi P, Cheng C. Potential therapies targeting nuclear metabolic regulation in cancer. MedComm (Beijing) 2023; 4:e421. [PMID: 38034101 PMCID: PMC10685089 DOI: 10.1002/mco2.421] [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: 05/09/2023] [Revised: 09/28/2023] [Accepted: 10/12/2023] [Indexed: 12/02/2023] Open
Abstract
The interplay between genetic alterations and metabolic dysregulation is increasingly recognized as a pivotal axis in cancer pathogenesis. Both elements are mutually reinforcing, thereby expediting the ontogeny and progression of malignant neoplasms. Intriguingly, recent findings have highlighted the translocation of metabolites and metabolic enzymes from the cytoplasm into the nuclear compartment, where they appear to be intimately associated with tumor cell proliferation. Despite these advancements, significant gaps persist in our understanding of their specific roles within the nuclear milieu, their modulatory effects on gene transcription and cellular proliferation, and the intricacies of their coordination with the genomic landscape. In this comprehensive review, we endeavor to elucidate the regulatory landscape of metabolic signaling within the nuclear domain, namely nuclear metabolic signaling involving metabolites and metabolic enzymes. We explore the roles and molecular mechanisms through which metabolic flux and enzymatic activity impact critical nuclear processes, including epigenetic modulation, DNA damage repair, and gene expression regulation. In conclusion, we underscore the paramount significance of nuclear metabolic signaling in cancer biology and enumerate potential therapeutic targets, associated pharmacological interventions, and implications for clinical applications. Importantly, these emergent findings not only augment our conceptual understanding of tumoral metabolism but also herald the potential for innovative therapeutic paradigms targeting the metabolism-genome transcriptional axis.
Collapse
Affiliation(s)
- Yanjie Chen
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jie Xu
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaoyi Liu
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Linlin Guo
- Department of Microbiology and ImmunologyThe Indiana University School of MedicineIndianapolisIndianaUSA
| | - Ping Yi
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Chunming Cheng
- Department of Radiation OncologyJames Comprehensive Cancer Center and College of Medicine at The Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
17
|
Wang L, Du C, Jiang B, Chen L, Wang Z. Adjusting the dose of traditional drugs combined with immunotherapy: reshaping the immune microenvironment in lung cancer. Front Immunol 2023; 14:1256740. [PMID: 37901223 PMCID: PMC10600379 DOI: 10.3389/fimmu.2023.1256740] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/05/2023] [Indexed: 10/31/2023] Open
Abstract
Immunotherapy is currently the most promising clinical treatment for lung cancer, not only revolutionizing second-line therapy but now also approved for first-line treatment. However, its clinical efficiency is not high and not all patients benefit from it. Thus, finding the best combination strategy to expand anti-PD-1/PD-L1-based immunotherapy is now a hot research topic. The conventional use of chemotherapeutic drugs and targeted drugs inevitably leads to resistance, toxic side effects and other problems. Recent research, however, suggests that by adjusting the dosage of drugs and blocking the activation of mutational mechanisms that depend on acquired resistance, it is possible to reduce toxic side effects, activate immune cells, and reshape the immune microenvironment of lung cancer. Here, we discuss the effects of different chemotherapeutic drugs and targeted drugs on the immune microenvironment. We explore the effects of adjusting the dosing sequence and timing, and the mechanisms of such responses, and show how the effectiveness and reliability of combined immunotherapy provide improved treatment outcomes.
Collapse
Affiliation(s)
- Linlin Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Changqi Du
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Bing Jiang
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Lin Chen
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zibing Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| |
Collapse
|
18
|
Zhou Y, Zhang A, Fang C, Yuan L, Shao A, Xu Y, Zhou D. Oxidative stress in pituitary neuroendocrine tumors: Affecting the tumor microenvironment and becoming a new target for pituitary neuroendocrine tumor therapy. CNS Neurosci Ther 2023; 29:2744-2759. [PMID: 37341156 PMCID: PMC10493678 DOI: 10.1111/cns.14315] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/22/2023] Open
Abstract
Pituitary adenomas (PAs), or pituitary neuroendocrine tumors (PitNETs), are commonly found in the anterior pituitary gland. Although the majority of PitNETs are benign and stable, several tumors have malignant characteristics. The tumor microenvironment (TME) plays an important role in the process of tumorigenesis and is composed of several types of cells. Various cells in the TME are significantly affected by oxidative stress. It has been reported that immunotherapeutic strategies have good effects in several cancers. However, the clinical potential of immunotherapies in PitNETs has not yet been fully discussed. Oxidative stress can regulate PitNET cells and immune cells in the TME, thus affecting the immune status of the TME of PitNETs. Therefore, modulation of oxidative stress-regulated immune cells using a combination of several agents and the immune system to suppress PitNETs is a promising therapeutic direction. In this review, we systematically analyzed the oxidative stress process within PitNET cells and various immune cells to elucidate the potential value of immunotherapy.
Collapse
Affiliation(s)
- Yuhang Zhou
- The First Clinical Medical CollegeHeilongjiang University of Chinese MedicineHarbinChina
- Health Management CenterTongde Hospital of Zhejiang ProvinceHangzhouChina
| | - Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Ling Yuan
- School of Public Health, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Yuanzhi Xu
- Department of Neurosurgery, Huashan Hospital, School of MedicineFudan UniversityShanghaiChina
| | - Danyang Zhou
- Health Management CenterTongde Hospital of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
19
|
Zhang X, Ma L, Wang J. Cross-Regulation Between Redox and Epigenetic Systems in Tumorigenesis: Molecular Mechanisms and Clinical Applications. Antioxid Redox Signal 2023; 39:445-471. [PMID: 37265163 DOI: 10.1089/ars.2023.0253] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Significance: Redox and epigenetics are two important regulatory processes of cell physiological functions. The cross-regulation between these processes has critical effects on the occurrence and development of various types of tumors. Recent Advances: The core factor that influences redox balance is reactive oxygen species (ROS) generation. The ROS functions as a double-edged sword in tumors: Low levels of ROS promote tumors, whereas excessive ROS induces various forms of tumor cell death, including apoptosis and ferroptosis as well as necroptosis and pyroptosis. Many studies have shown that the redox balance is influenced by epigenetic mechanisms such as DNA methylation, histone modification, chromatin remodeling, non-coding RNAs (microRNA, long non-coding RNA, and circular RNA), and RNA N6-methyladenosine modification. Several oxidizing or reducing substances also affect the epigenetic state. Critical Issues: In this review, we summarize research on the cross-regulation between redox and epigenetics in cancer and discuss the relevant molecular mechanisms. We also discuss the current research on the clinical applications. Future Directions: Future research can use high-throughput methods to analyze the molecular mechanisms of the cross-regulation between redox and epigenetics using both in vitro and in vivo models in more detail, elucidate regulatory mechanisms, and provide guidance for clinical treatment. Antioxid. Redox Signal. 39, 445-471.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Lifang Ma
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jiayi Wang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
20
|
Herzog BH, Baer JM, Borcherding N, Kingston NL, Belle JI, Knolhoff BL, Hogg GD, Ahmad F, Kang LI, Petrone J, Lin CY, Govindan R, DeNardo DG. Tumor-associated fibrosis impairs immune surveillance and response to immune checkpoint blockade in non-small cell lung cancer. Sci Transl Med 2023; 15:eadh8005. [PMID: 37285399 DOI: 10.1126/scitranslmed.adh8005] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/17/2023] [Indexed: 06/09/2023]
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related deaths. Immune checkpoint blockade has improved survival for many patients with NSCLC, but most fail to obtain long-term benefit. Understanding the factors leading to reduced immune surveillance in NSCLC is critical in improving patient outcomes. Here, we show that human NSCLC harbors large amounts of fibrosis that correlates with reduced T cell infiltration. In murine NSCLC models, the induction of fibrosis led to increased lung cancer progression, impaired T cell immune surveillance, and failure of immune checkpoint blockade efficacy. Associated with these changes, we observed that fibrosis leads to numerically and functionally impaired dendritic cells and altered macrophage phenotypes that likely contribute to immunosuppression. Within cancer-associated fibroblasts, distinct changes within the Col13a1-expressing population suggest that these cells produce chemokines to recruit macrophages and regulatory T cells while limiting recruitment of dendritic cells and T cells. Targeting fibrosis through transforming growth factor-β receptor signaling overcame the effects of fibrosis to enhance T cell responses and improved the efficacy of immune checkpoint blockade but only in the context of chemotherapy. Together, these data suggest that fibrosis in NSCLC leads to reduced immune surveillance and poor responsiveness to checkpoint blockade and highlight antifibrotic therapies as a candidate strategy to overcome immunotherapeutic resistance.
Collapse
Affiliation(s)
- Brett H Herzog
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John M Baer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Natalie L Kingston
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jad I Belle
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brett L Knolhoff
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Graham D Hogg
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Faiz Ahmad
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Liang-I Kang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jessica Petrone
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ramaswamy Govindan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David G DeNardo
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
21
|
Renaud E, Ricordel C, Corre R, Leveiller G, Gadby F, Babey H, Annic J, Lucia F, Bourbonne V, Robinet G, Descourt R, Orione C, Quéré G, Geier M. Pembrolizumab plus pemetrexed-carboplatin combination in first-line treatment of advanced non-squamous non-small cell lung cancer: a multicenter real-life study (CAP29). Transl Lung Cancer Res 2023; 12:266-276. [PMID: 36895931 PMCID: PMC9989798 DOI: 10.21037/tlcr-22-556] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 01/11/2023] [Indexed: 03/01/2023]
Abstract
Background Pembrolizumab combined with chemotherapy is now first-line standard of care in advanced non-small cell lung cancer. This real-life study aimed to assess efficacy and safety of carboplatin-pemetrexed plus pembrolizumab in advanced non-squamous non-small cell lung cancer. Methods CAP29 is a retrospective, observational, multicenter real-life study conducted in 6 French centers. We evaluated efficacy of first-line setting chemotherapy plus pembrolizumab (November 2019 to September 2020) in advanced (stage III-IV) non-squamous non-small cell lung cancer patients without targetable alterations. Primary endpoint was progression-free survival. Secondary endpoints were overall survival, objective response rate and safety. Results With a median follow-up of 4.5 months (0 to 22 months), a total of 121 patients were included. Baseline characteristics were: median age of 59.8 years with 7.4% ≥75 years, 58.7% of males, 91.8% PS 0-1, 87.6% of stage IV with ≥3 metastatic sites in 62% of cases. Patients had brain and liver metastases in 24% and 15.7% of cases, respectively. PD-L1 was <1% (44.6%), 1-49% (28.1%) and ≥50% (21.5%). Median progression-free survival and overall survival achieved 9 and 20.6 months, respectively. Objective response rate was 63.7% with 7 prolonged complete responses. Survival benefit seemed to be correlated with PD-L1 expression. Brain and liver metastases were not statistically associated with decreased overall survival. Most common adverse events were asthenia (76%), anemia (61.2%), nausea (53.7%), decreased appetite (37.2%) and liver cytolysis (34.7%). Renal and hepatic disorders were the main causes of pemetrexed discontinuation. Grade 3-4 adverse events concerned 17.5% of patients. Two treatment-related deaths were reported. Conclusions First-line pembrolizumab plus chemotherapy confirmed real-life efficacy for patients with advanced non-squamous non-small cell lung cancer. With median progression-free survival and overall survival of 9.0 and 20.6 months, respectively and no new safety signal, our real-life data are very close to results provided by clinical trials, confirming the benefit and the manageable toxicity profile of this combination.
Collapse
Affiliation(s)
- Emmanuelle Renaud
- Department of Medical Oncology, CHRU Morvan, University Hospital of Brest, Brest, France
| | - Charles Ricordel
- Department of Pulmonary Diseases, CHU Pontchaillou, University Hospital of Rennes, Rennes, France
| | - Romain Corre
- Department of Pulmonary Diseases, CH Cornouaille, Quimper, France
| | | | - Florence Gadby
- Department of Pulmonary Diseases, Hospital of Morlaix, Morlaix, France
| | - Hélène Babey
- Department of Medical Oncology, Hospital of Landerneau, Landerneau, France
| | - Josselin Annic
- Department of Medical Oncology, CHRU Morvan, University Hospital of Brest, Brest, France
| | - François Lucia
- Department of Radiation Oncology, CHRU Morvan, University Hospital of Brest, Brest, France
| | - Vincent Bourbonne
- Department of Radiation Oncology, CHRU Morvan, University Hospital of Brest, Brest, France
| | - Gilles Robinet
- Department of Medical Oncology, CHRU Morvan, University Hospital of Brest, Brest, France
| | - Renaud Descourt
- Department of Medical Oncology, CHRU Morvan, University Hospital of Brest, Brest, France
| | - Charles Orione
- Department of Pulmonary Diseases, CHRU Cavale Blanche University Hospital of Brest, Brest, France
| | - Gilles Quéré
- Department of Medical Oncology, CHRU Morvan, University Hospital of Brest, Brest, France
| | - Margaux Geier
- Department of Medical Oncology, CHRU Morvan, University Hospital of Brest, Brest, France
| |
Collapse
|
22
|
Zhang X, Zhang J, Liu P, Wang J, Zhao K, Zhu Z, Gu K, Zhao W. Immunotherapy progress and clinical strategy of unresectable locally advanced non-small cell lung cancer. Front Oncol 2023; 13:1022042. [PMID: 36845690 PMCID: PMC9945126 DOI: 10.3389/fonc.2023.1022042] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
Non-small cell lung cancer negative for actionable molecular markers entered the splendid era of immunotherapy. This review aims to provide an evidence-based summary for immunotherapy for unresectable locally advanced non-small cell lung cancer, and references for clinical strategies of immunotherapy. Through literature review, the standard treatment for unresectable locally advanced non-small cell lung cancer should be radical concurrent radiotherapy and chemotherapy followed by consolidation immunotherapy. However, the efficacy of concurrent radiotherapy, chemotherapy combined with immunotherapy has not been improved, and its safety should be further validated. It is believed that induction immunotherapy plus concurrent radiotherapy and chemotherapy plus consolidation immunotherapy is promising. In clinical practice, the delineation of radiotherapy target should be relatively small. Pemetrexed combined with PD-1 inhibitor induces the strongest immunogenicity in chemotherapy, which is suggested by preclinical pathway study. Although there is no significant difference between PD1 and PD1 for effect, PD-L1 inhibitor is better in the combination treatment of radiotherapy which presents significantly less adverse events.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianguo Zhang
- Department of Oncology, Lu‘an civily Hospital, Anhui, Lu’an, China
| | - Peiyi Liu
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China
| | - Juan Wang
- Department of Oncology, The Second People’s Hospital of Kashgar, Kashgar, China
| | - Kuaile Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kangsheng Gu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Weixin Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Shi MY, Liu HG, Chen XH, Tian Y, Chen ZN, Wang K. The application basis of immuno-checkpoint inhibitors combined with chemotherapy in cancer treatment. Front Immunol 2023; 13:1088886. [PMID: 36703971 PMCID: PMC9871553 DOI: 10.3389/fimmu.2022.1088886] [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: 11/03/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Immuno-checkpoint inhibitors (ICIs) bring a promising prospect for patients with cancers, which restrains the growth of tumor cells by enhancing anti-tumor activity. Nevertheless, not all patients benefit from the administration of ICIs monotherapy. The partial response or resistance to ICIs is mainly due to the complex and heterogenous tumor microenvironment (TME). The combined therapy is necessary for improving the efficacy of tumor treatment. Chemotherapy is reported not only to kill tumor cells directly, but also to stimulate effective anti-tumor immune responses. Several combined therapies of ICIs and chemotherapeutic agents have been approved for the first-line treatment of cancers, including PD-1/PD-L1 inhibitors. This review summarizes the potential mechanisms of the combined therapy of ICIs and chemotherapeutic agents in inducing immunogenic cell death (ICD) and reprogramming TME, and elucidates the possible anti-tumor effects of combined therapy from the perspective of metabolic reprogramming and microbiome reprogramming.
Collapse
Affiliation(s)
| | | | | | | | | | - Ke Wang
- *Correspondence: Ke Wang, ; Zhi-Nan Chen,
| |
Collapse
|
24
|
Liu BW, Shang QX, Yang YS, Chen LQ. Efficacy and safety of PD-1/PD-L1 inhibitor combined with chemotherapy versus chemotherapy alone in the treatment of advanced gastric or gastroesophageal junction adenocarcinoma: a systematic review and meta-analysis. Front Oncol 2023; 13:1077675. [PMID: 37114136 PMCID: PMC10129365 DOI: 10.3389/fonc.2023.1077675] [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: 11/24/2022] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Background There is increasing evidence that immunotherapy (programmed cell death-1 (PD-1) inhibitor) combined with chemotherapy is superior to chemotherapy alone in neoadjuvant therapy for patients with previously untreated, unresectable advanced, or metastatic esophageal adenocarcinoma (EAC)/gastric/gastroesophageal junction adenocarcinoma (GEA). However, the results of recent studies have been contradictory. Therefore, the aim of this article is to evaluate the efficacy and safety of PD-1 inhibitors combined with chemotherapy in neoadjuvant therapy through meta-analysis. Method We comprehensively reviewed the literature and clinical randomized controlled trials (RCTs) by February 2022 by searching Medical Subject Headings (MeSH) and keywords such as "esophageal adenocarcinoma" or "immunotherapy" in several databases, including the Embase, Cochrane, PubMed, and ClinicalTrials.gov websites. Two authors independently selected studies, extracted data, and assessed the risk of bias and quality of evidence by using standardized Cochrane Methods procedures. The primary outcomes were 1-year overall survival (OS) and 1-year progression-free survival (PFS), estimated by calculating the 95% confidence interval (CI) for the combined odds ratio (OR) and hazard ratio (HR). Secondary outcomes estimated using OR were disease objective response rate (DORR) and incidence of adverse events. Results Four RCTs with a total of 3,013 patients researching the efficacy of immunotherapy plus chemotherapy versus chemotherapy alone on gastrointestinal cancer were included in this meta-analysis. The results showed that immune checkpoint inhibitor plus chemotherapy treatment was associated with an increased risk of PFS (HR = 0.76 [95% CI: 0.70-0.83]; p < 0.001), OS (HR = 0.81 [95% CI: 0.74-0.89]; p < 0.001), and DORR (relative ratio (RR) = 1.31 [95% CI: 1.19-1.44]; p < 0.0001) when compared with chemotherapy alone in advanced, unresectable, and metastatic EAC/GEA. However, immunotherapy combined with chemotherapy increased the incidence of adverse reactions such as alanine aminotransferase elevation (OR = 1.55 [95% CI: 1.17-2.07]; p = 0.003) and palmar-plantar erythrodysesthesia (PPE) syndrome (OR = 1.30 [95% CI: 1.05-1.63]; p = 0.02). Nausea (OR = 1.24 [95% CI: 1.07-1.44]; p = 0.005) and white blood cell count decreased (OR = 1.40 [95% CI: 1.13-1.73]; p = 0.002), and so on. Fortunately, toxicities were within acceptable limits. Meanwhile, for patients with a combined positive score (CPS) ≥1, compared with chemotherapy alone, immunotherapy combined with chemotherapy had a better overall survival rate (HR = 0.81 [95% CI: 0.73-0.90]; p = 0.0001). Conclusion Our study shows that immunotherapy plus chemotherapy has an obvious benefit for patients with previously untreated, unresectable advanced, or metastatic EAC/GEA when compared with chemotherapy alone. However, a high risk of adverse reactions may occur during immunotherapy plus chemotherapy, and more studies focusing on the treatment strategies of untreated, unresectable advanced, or metastatic EAC/GEA are warranted. Systematic review registration www.crd.york.ac.uk, identifier CRD42022319434.
Collapse
|
25
|
Zhao Y, Wang Z, Shi X, Liu T, Yu W, Ren X, Zhao H. Effect of Chemotherapeutics on In Vitro Immune Checkpoint Expression in Non-Small Cell Lung Cancer. Technol Cancer Res Treat 2023; 22:15330338231202307. [PMID: 37728201 PMCID: PMC10515539 DOI: 10.1177/15330338231202307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/03/2023] [Accepted: 07/28/2023] [Indexed: 09/21/2023] Open
Abstract
Objectives: Immune checkpoint (ICP) expression in tumor cells could directly or indirectly affect the results of immunotherapy. ICP ligands on tumor cells usually bind their immune cell receptors to inhibit the activity, resulting in tumor immune escape. Thus, the purpose of this study was to ascertain the impact of various chemotherapeutic drugs on ICP expression in non-small cell lung cancer (NSCLC) cell lines with different pathological subtypes to provide a basis for the development of a superior regimen of chemotherapy combined with ICP blockade. Methods: Several first-line chemotherapy agents (cisplatin, carboplatin, paclitaxel, gemcitabine, vinorelbine, and pemetrexed) were selected to treat different NSCLC cell lines (squamous carcinoma H1703, adenocarcinoma A549, and large cell cancer H460) for 72 hours, and then the changes in ICP expression in the tumor cells were observed through flow cytometry. Results: Cisplatin, carboplatin, and paclitaxel upregulated the expressions of programmed cell death ligand 1 (PD-L1) and programmed cell death ligand 2 (PD-L2) in A549 and H460 cell lines. Meanwhile, vinorelbine and pemetrexed upregulated PD-L1 and PD-L2 in H1703, A549, and H460 cell lines. Paclitaxel, gemcitabine, vinorelbine, and pemetrexed significantly upregulated the expressions of both galectin-9 and high-mobility group box protein 1 (HMGB1) in the A549 cell line. Cisplatin and paclitaxel significantly upregulated the expressions of major histocompatibility complex-II (MHC-II), galectin-3, α-synuclein, and fibrinogen-like protein 1 (FGL1) in A549 and H460 cell lines. In addition, cisplatin and vinorelbine significantly upregulated the expressions of both CD155 and CD112 in the H460 cell line. Vinorelbine upregulated MHC-I in all three cell lines. Conclusion: Chemotherapy agents have different effects on the expression of ICP ligands in tumor cells with different pathological types, and this may affect the efficacy of combined immunotherapy. These results provide a theoretical basis for further selection and optimization of the combination of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Yu Zhao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Zhe Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiuhuan Shi
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Ting Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Wenwen Yu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Haihe Laboratory of Cell Ecosystem, Tianjin, China
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hua Zhao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Haihe Laboratory of Cell Ecosystem, Tianjin, China
| |
Collapse
|
26
|
Hyroššová P, Milošević M, Škoda J, Vachtenheim Jr J, Rohlena J, Rohlenová K. Effects of metabolic cancer therapy on tumor microenvironment. Front Oncol 2022; 12:1046630. [PMID: 36582801 PMCID: PMC9793001 DOI: 10.3389/fonc.2022.1046630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Targeting tumor metabolism for cancer therapy is an old strategy. In fact, historically the first effective cancer therapeutics were directed at nucleotide metabolism. The spectrum of metabolic drugs considered in cancer increases rapidly - clinical trials are in progress for agents directed at glycolysis, oxidative phosphorylation, glutaminolysis and several others. These pathways are essential for cancer cell proliferation and redox homeostasis, but are also required, to various degrees, in other cell types present in the tumor microenvironment, including immune cells, endothelial cells and fibroblasts. How metabolism-targeted treatments impact these tumor-associated cell types is not fully understood, even though their response may co-determine the overall effectivity of therapy. Indeed, the metabolic dependencies of stromal cells have been overlooked for a long time. Therefore, it is important that metabolic therapy is considered in the context of tumor microenvironment, as understanding the metabolic vulnerabilities of both cancer and stromal cells can guide new treatment concepts and help better understand treatment resistance. In this review we discuss recent findings covering the impact of metabolic interventions on cellular components of the tumor microenvironment and their implications for metabolic cancer therapy.
Collapse
Affiliation(s)
- Petra Hyroššová
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| | - Mirko Milošević
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Josef Škoda
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| | - Jiří Vachtenheim Jr
- 3rd Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Jakub Rohlena
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| | - Kateřina Rohlenová
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
27
|
The Potential Mechanism of HDAC1-Catalyzed Histone Crotonylation of Caspase-1 in Nonsmall Cell Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5049116. [PMID: 35958929 PMCID: PMC9363190 DOI: 10.1155/2022/5049116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022]
Abstract
Nonsmall cell lung cancer (NSCLC) is a predominant subtype of lung cancer and accounts for over 80% of all lung cancer cases. The resistance to pemetrexed (PEM) is frequently occurred and severely affects the NSCLC therapy. Proteomic analysis of histones indicated that the histone deacetylase 1 (HDAC1) complex could hydrolyze lysine crotonylation on histone3 (H3K18cr), affecting epigenetic regulation in cancers. However, the effect of HDAC1-mediated H3K18cr on the PEM resistance of NSCLC is still unclear. Here, we aimed to explore the function of HDAC1-mediated H3K18cr in NSCLC PEM resistance. The expression of HDAC1 was upregulated in clinical NSCLC tissues and cell lines and correlated with the poor prognosis of NSCLC samples. We constructed the PEM-resistant NSCLC cell lines, and the depletion of HDAC1 remarkably reduced the viability of the cells. The proliferation of PEM-resistant NSCLC cells was decreased by HDAC1 knockdown, and the IC50 of PEM was repressed by the silencing of HDAC1 in the cells. Mechanically, we identified the enrichment of HDAC1 on the promoter of caspase-1 in PEM-resistant NSCLC cells. The depletion of HDAC1 inhibited the enrichment of histone H3K18cr and RNA polymerase II (RNA pol II) on the caspase-1 promoter in the cells. The expression of caspase-1 was suppressed by HDAC1 knockdown. The knockdown of HDAC1 reduced proliferation of PEM-resistant NSCLC cells, in which caspase-1 or GSDMD depletion reversed the effect. Clinically, the HDAC1 expression was negatively associated with caspase-1 and GSDMD in clinical NSCLC tissues, while caspase-1 and GSDMD expression was positively correlated in the samples. Therefore, we concluded that HDAC1-catalyzed histone crotonylation of caspase-1 modulates PEM sensitivity of NSCLC by targeting GSDMD.
Collapse
|
28
|
Principe DR, Kamath SD, Korc M, Munshi HG. The immune modifying effects of chemotherapy and advances in chemo-immunotherapy. Pharmacol Ther 2022; 236:108111. [PMID: 35016920 PMCID: PMC9271143 DOI: 10.1016/j.pharmthera.2022.108111] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/06/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment paradigm for several malignancies. While the use of single-agent or combined ICIs has achieved acceptable disease control rates in a variety of solid tumors, such approaches have yet to show substantial therapeutic efficacy in select difficult-to-treat cancer types. Recently, select chemotherapy regimens are emerging as extensive modifiers of the tumor microenvironment, leading to the reprogramming of local immune responses. Accordingly, data is now emerging to suggest that certain anti-neoplastic agents modulate various immune cell processes, most notably the cross-presentation of tumor antigens, leukocyte trafficking, and cytokine biosynthesis. As such, the combination of ICIs and cytotoxic chemotherapy are beginning to show promise in many cancers that have long been considered poorly responsive to ICI-based immunotherapy. Here, we discuss past and present attempts to advance chemo-immunotherapy in these difficult-to-treat cancer histologies, mechanisms through which select chemotherapies modify tumor immunogenicity, as well as important considerations when designing such approaches to maximize efficacy and improve therapeutic response rates.
Collapse
Affiliation(s)
- Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, USA; Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, USA.
| | - Suneel D Kamath
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Murray Korc
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Hidayatullah G Munshi
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
29
|
Interaction between baseline HBV loads and the prognosis of patients with HCC receiving anti-PD-1 in combination with antiangiogenic therapy undergoing concurrent TAF prophylaxis. BMC Infect Dis 2022; 22:614. [PMID: 35836207 PMCID: PMC9284788 DOI: 10.1186/s12879-022-07602-0] [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: 04/21/2022] [Accepted: 07/07/2022] [Indexed: 12/24/2022] Open
Abstract
Background A high baseline hepatitis B virus (HBV) load has always been listed as an exclusion criterion for programmed cell death-1 (PD-1) inhibitor-associated therapy in clinical trials, as the interaction between HBV load and anti-PD-1/PD-L1 therapy with anti HBV therapy remains controversial. Methods We retrospectively enrolled 70 unresectable HCC patients who were seropositive for HBsAg and accepted tenofovir alafenamide fumarate (TAF) therapy before anti-PD-1 in combination with an antiangiogenic treatment. Patients were divided into a low HBV DNA group (≤ 2000 IU/ml) and a high HBV DNA group (> 2000 IU/ml) according to the baseline HBV DNA levels. Tumour response and progression-free survival (PFS) were compared, and univariate and multivariate Cox analyses were performed to identify potential risk factors for PFS. The incidences of HBV reactivation and HBV-associated hepatitis were also recorded. Results 48 patients were assigned to the low group and the remaining 22 patients were assigned to the high group. The objective response rates (ORRs), disease control rates (DCRs), and PFS between the two groups showed no significant difference (P = 0.761, 0.552, and 0.784, respectively). The results of Cox analyses revealed that there was no relationship between baseline HBV load and PFS. Additionally, HBV reactivation occurred in only 2 patients (2.9%), and no patient experienced HBV-related hepatic impairment when given a continuous TAF treatment. Conclusions Baseline HBV loads do not affect the prognosis of HCC patients receiving anti-PD-1 in combination with an antiangiogenic therapy, while PD-1 inhibitors do not aggravate HBV reactivation and hepatic impairment in patients simultaneously subjected to TAF prophylaxis. Baseline HBV loads do not affect the prognosis of HCC patients receiving anti-PD-1 in combination with antiangiogenic therapy. Besides, PD-1 inhibitors do not aggravate HBV reactivation and hepatic impairment undergoing concurrent TAF prophylaxis.
Collapse
|
30
|
Computed Tomography Image under Artificial Intelligence Algorithm to Evaluate the Nursing and Treatment Effect of Pemetrexed Combined Platinum-Based Chemotherapy on Elderly Lung Cancer. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:2574451. [PMID: 35800237 PMCID: PMC9192264 DOI: 10.1155/2022/2574451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/03/2022] [Accepted: 05/16/2022] [Indexed: 11/17/2022]
Abstract
This study was to evaluate the clinical efficacy of pemetrexed combined with platinum-based chemotherapy in the treatment of elderly lung cancer using electronic computed tomography (CT) images based on artificial intelligence algorithms. In this study, 80 elderly patients with lung cancer treated were selected and randomly divided into two groups: patients treated with pemetrexed combined with cisplatin were included in the pemetrexed group and patients treated with docetaxel combined with cisplatin were included in the docetaxel group, with 40 cases in each group. The DenseNet network was compared with the Let Net-5 and ResNet model and applied to the CT images of 80 elderly patients with lung cancer. The diagnosis accuracy of the DenseNet network (97.4%) was higher than that of the Let Net-5 network (80.1%) and ResNet model (95.5%). Carcinoembryonic antigen (CEA), cytokeratin fragment antigen 21–1 (CYFRA 21–1), and squamous cell-associated antigen (SCC) after chemotherapy in the pemetrexed group and docetaxel group were all lower than those before chemotherapy, showing statistically obvious differences (P < 0.05). The satisfaction degree of nursing care in the pemetrexed group (92.67%) was significantly higher than that in the docetaxel group (85.62%), and the difference was statistically significant (P < 0.05). Adverse reactions such as fatigue, diarrhea, and neutrophils in the pemetrexed group were lower than those in the docetaxel group, and the difference was statistically great (P < 0.05). The DenseNet convolutional neural network has high diagnostic accuracy; methotrexate combined with platinum chemotherapy can improve the chemotherapy effect in elderly patients with lung cancer, with low degree of adverse reactions and good overall tolerance, which can be used as the first-line treatment for elderly patients with lung cancer.
Collapse
|
31
|
miR-6077 promotes cisplatin/pemetrexed resistance in lung adenocarcinoma via CDKN1A/cell cycle arrest and KEAP1/ferroptosis pathways. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 28:366-386. [PMID: 35505963 PMCID: PMC9035384 DOI: 10.1016/j.omtn.2022.03.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/27/2022] [Indexed: 01/18/2023]
Abstract
Lung adenocarcinoma (LUAD) is one of the most common malignancies worldwide. Combination chemotherapy with cisplatin (CDDP) plus pemetrexed (PEM) remains the predominant therapeutic regimen; however, chemoresistance greatly limits its curative potential. Here, through CRISPR-Cas9 screening, we identified miR-6077 as a key driver of CDDP/PEM resistance in LUAD. Functional experiments verified that ectopic overexpression of miR-6077 desensitized LUAD cells to CDDP/PEM in both cell lines and patient-derived xenograft models. Through RNA sequencing in cells and single-cell sequencing of samples from patients with CDDP/PEM treatments, we observed CDDP/PEM-induced upregulation of CDKN1A and KEAP1, which in turn activated cell-cycle arrest and ferroptosis, respectively, thus leading to cell death. Through miRNA pull-down, we identified and validated that miR-6077 targets CDKN1A and KEAP1. Furthermore, we demonstrated that miR-6077 protects LUAD cells from cell death induced by CDDP/PEM via CDKN1A-CDK1-mediated cell-cycle arrest and KEAP1-NRF2-SLC7A11/NQO1-mediated ferroptosis, thus resulting in chemoresistance in multiple LUAD cells both in vitro and in vivo. Moreover, we found that GMDS-AS1 and LINC01128 sensitized LUAD cells to CDDP/PEM by sponging miR-6077. Collectively, these results imply the critical role of miR-6077 in LUAD’s sensitivity to CDDP/PEM, thus providing a novel therapeutic strategy for overcoming chemoresistance in clinical practice.
Collapse
|
32
|
Lin CW, Chen CC, Huang WY, Chen YY, Chen ST, Chou HW, Hung CM, Chen WJ, Lu CS, Nian SX, Chen SG, Chang HW, Chang VH, Liu LY, Kuo ML, Chang SC. Restoring Pro-healing/remodeling- associated M2a/c Macrophages using ON101 Accelerates Diabetic Wound Healing. JID INNOVATIONS 2022; 2:100138. [PMID: 36017415 PMCID: PMC9396230 DOI: 10.1016/j.xjidi.2022.100138] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 01/05/2023] Open
Abstract
Diabetic wounds exhibit chronic inflammation and delayed tissue proliferation or remodeling, mainly owing to prolonged proinflammatory (M1) macrophage activity and defects in transition to prohealing/proremodeling (M2a/M2c; CD206+ and/or CD163+) macrophages. We found that topical treatment with ON101, a plant-based potential therapeutic for diabetic foot ulcers, increased M2c-like (CD163+ and CD206+) cells and suppressed M1-like cells, altering the inflammatory gene profile in a diabetic mouse model compared with that in the controls. An in vitro macrophage-polarizing model revealed that ON101 directly suppressed CD80+ and CD86+ M1-macrophage polarization and M1-associated proinflammatory cytokines at both protein and transcriptional levels. Notably, conditioned medium collected from ON101-treated M1 macrophages reversed the M1-conditioned medium‒mediated suppression of CD206+ macrophages. Furthermore, conditioned medium from ON101-treated adipocyte progenitor cells significantly promoted CD206+ and CD163+ macrophages but strongly inhibited M1-like cells. ON101 treatment also stimulated the expression of GCSF and CXCL3 genes in human adipocyte progenitor cells. Interestingly, treatment with recombinant GCSF protein enhanced both CD206+ and CD163+ M2 markers, whereas CXCL3 treatment only stimulated CD163+ M2 macrophages. Depletion of cutaneous M2 macrophages inhibited ON101-induced diabetic wound healing. Thus, ON101 directly suppressed M1 macrophages and facilitated the GCSF- and CXCL3-mediated transition from M1 to M2 macrophages, lowering inflammation and leading to faster diabetic wound healing.
Collapse
Affiliation(s)
| | - Chih-Chiang Chen
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Dermatology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | | | | | | | | | | | | | - Chia-Sing Lu
- NTU YongLin Institute of Health, National Taiwan University, Taipei, Taiwan
| | - Shi-Xin Nian
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shyi-Gen Chen
- Oneness Biotech Co., Ltd., Taipei, Taiwan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsuen-Wen Chang
- TMU Laboratory Animal Center, Office of Research and Development, Taipei Medical University, Taipei, Taiwan
| | - Vincent H.S. Chang
- TMU Laboratory Animal Center, Office of Research and Development, Taipei Medical University, Taipei, Taiwan
- Department of Physiology, School of Medicine, Taipei Medical University, Taipei, Taiwan
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Li-Ying Liu
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | - Shun-Cheng Chang
- Division of Plastic Surgery, Integrated Burn & Wound Care Center, Department of Surgery, Shuang-Ho Hospital; Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Correspondence: Shun-Cheng Chang, Division of Plastic Surgery, Integrated Burn & Wound Care Center, Department of Surgery, Shuang-Ho Hospital; Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Number 291, Zhongzheng Road, Zhonghe District, New Taipei City 235, Taiwan.
| |
Collapse
|
33
|
Yao Q, Zhang X, Chen D. Emerging Roles and Mechanisms of lncRNA FOXD3-AS1 in Human Diseases. Front Oncol 2022; 12:848296. [PMID: 35280790 PMCID: PMC8914342 DOI: 10.3389/fonc.2022.848296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/01/2022] [Indexed: 01/02/2023] Open
Abstract
Numerous long noncoding RNAs (lncRNAs) have been identified as powerful regulators of human diseases. The lncRNA FOXD3-AS1 is a novel lncRNA that was recently shown to exert imperative roles in the initialization and progression of several diseases. Emerging studies have shown aberrant expression of FOXD3-AS1 and close correlation with pathophysiological traits of numerous diseases, particularly cancers. More importantly, FOXD3-AS1 was also found to ubiquitously impact a range of biological functions. This study aims to summarize the expression, associated clinicopathological features, major functions and molecular mechanisms of FOXD3-AS1 in human diseases and to explore its possible clinical applications.
Collapse
Affiliation(s)
- Qinfan Yao
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Xiuyuan Zhang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Dajin Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- *Correspondence: Dajin Chen,
| |
Collapse
|
34
|
Cioce M, Canino C, Pass H, Blandino G, Strano S, Fazio VM. Arachidonic acid drives adaptive responses to chemotherapy-induced stress in malignant mesothelioma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:344. [PMID: 34727953 PMCID: PMC8561918 DOI: 10.1186/s13046-021-02118-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022]
Abstract
Background High resistance to therapy and poor prognosis characterizes malignant pleural mesothelioma (MPM). In fact, the current lines of treatment, based on platinum and pemetrexed, have limited impact on the survival of MPM patients. Adaptive response to therapy-induced stress involves complex rearrangements of the MPM secretome, mediated by the acquisition of a senescence-associated-secretory-phenotype (SASP). This fuels the emergence of chemoresistant cell subpopulations, with specific gene expression traits and protumorigenic features. The SASP-driven rearrangement of MPM secretome takes days to weeks to occur. Thus, we have searched for early mediators of such adaptive process and focused on metabolites differentially released in mesothelioma vs mesothelial cell culture media, after treatment with pemetrexed. METHODS Mass spectrometry-based (LC/MS and GC/MS) identification of extracellular metabolites and unbiased statistical analysis were performed on the spent media of mesothelial and mesothelioma cell lines, at steady state and after a pulse with pharmacologically relevant doses of the drug. ELISA based evaluation of arachidonic acid (AA) levels and enzyme inhibition assays were used to explore the role of cPLA2 in AA release and that of LOX/COX-mediated processing of AA. QRT-PCR, flow cytometry analysis of ALDH expressing cells and 3D spheroid growth assays were employed to assess the role of AA at mediating chemoresistance features of MPM. ELISA based detection of p65 and IkBalpha were used to interrogate the NFkB pathway activation in AA-treated cells. RESULTS We first validated what is known or expected from the mechanism of action of the antifolate. Further, we found increased levels of PUFAs and, more specifically, arachidonic acid (AA), in the transformed cell lines treated with pemetrexed. We showed that pharmacologically relevant doses of AA tightly recapitulated the rearrangement of cell subpopulations and the gene expression changes happening in pemetrexed -treated cultures and related to chemoresistance. Further, we showed that release of AA following pemetrexed treatment was due to cPLA2 and that AA signaling impinged on NFkB activation and largely affected anchorage-independent, 3D growth and the resistance of the MPM 3D cultures to the drug. CONCLUSIONS AA is an early mediator of the adaptive response to pem in chemoresistant MPM and, possibly, other malignancies.
Collapse
Affiliation(s)
- Mario Cioce
- Department of Medicine, R.U. in Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128, Rome, Italy.
| | - Claudia Canino
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA.,Radiation Oncology Unit, UPMC Hillmann Cancer Center, San Pietro Hospital FBF, Rome, Italy
| | - Harvey Pass
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Sabrina Strano
- SAFU Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Vito Michele Fazio
- Department of Medicine, R.U. in Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128, Rome, Italy. .,Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133, Rome, Italy. .,Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy.
| |
Collapse
|
35
|
Chabanon RM, Rouanne M, Lord CJ, Soria JC, Pasero P, Postel-Vinay S. Targeting the DNA damage response in immuno-oncology: developments and opportunities. Nat Rev Cancer 2021; 21:701-717. [PMID: 34376827 DOI: 10.1038/s41568-021-00386-6] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
Immunotherapy has revolutionized cancer treatment and substantially improved patient outcome with regard to multiple tumour types. However, most patients still do not benefit from such therapies, notably because of the absence of pre-existing T cell infiltration. DNA damage response (DDR) deficiency has recently emerged as an important determinant of tumour immunogenicity. A growing body of evidence now supports the concept that DDR-targeted therapies can increase the antitumour immune response by (1) promoting antigenicity through increased mutability and genomic instability, (2) enhancing adjuvanticity through the activation of cytosolic immunity and immunogenic cell death and (3) favouring reactogenicity through the modulation of factors that control the tumour-immune cell synapse. In this Review, we discuss the interplay between the DDR and anticancer immunity and highlight how this dynamic interaction contributes to shaping tumour immunogenicity. We also review the most innovative preclinical approaches that could be used to investigate such effects, including recently developed ex vivo systems. Finally, we highlight the therapeutic opportunities presented by the exploitation of the DDR-anticancer immunity interplay, with a focus on those in early-phase clinical development.
Collapse
Affiliation(s)
- Roman M Chabanon
- ATIP-Avenir Group, Inserm Unit U981, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - Mathieu Rouanne
- Equipe Labellisée Ligue Nationale contre le Cancer, Inserm Unit U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Département d'Urologie, Hôpital Foch, Université Versailles-Saint-Quentin-en-Yvelines, Université Paris-Saclay, Suresnes, France
| | - Christopher J Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - Jean-Charles Soria
- Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médicine, Université Paris-Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Philippe Pasero
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Humaine, CNRS, Université de Montpellier, Montpellier, France
| | - Sophie Postel-Vinay
- ATIP-Avenir Group, Inserm Unit U981, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.
- Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France.
- Faculté de Médicine, Université Paris-Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France.
| |
Collapse
|
36
|
Nivolumab-induced membranous nephropathy in a patient with stage IV lung adenocarcinoma. CEN Case Rep 2021; 11:171-176. [PMID: 34524642 DOI: 10.1007/s13730-021-00645-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 09/05/2021] [Indexed: 01/03/2023] Open
Abstract
Immune check point inhibitors (ICIs) are now increasingly used for cancer therapy. At the same time, by activating the immune system, ICIs induce unique side effects, termed immune-related adverse events (irAEs). Renal irAEs, although uncommon, result in acute tubulointerstitial nephritis. Recently, because of an increase in ICI administration, renal irAEs, including glomerulonephritis, are being increasingly reported. A 69-year-old man presented with nephrotic syndrome after use of the ICI nivolumab. He underwent renal biopsy and was diagnosed with membranous nephropathy (MN) without acute tubulointerstitial nephritis. Immunofluorescence staining was negative for IgG4 and phospholipase A2 receptor (PLA2R), suggesting a malignancy-associated pattern. Oral glucocorticoid therapy was started as the standard treatment for irAEs, which resulted in complete remission of the nephrotic syndrome in 20 months. We suggest his MN was induced or accelerated by immune activation due to nivolumab. It means that ICIs possibly induce not only acute tubulointerstitial nephritis but also nephrotic syndrome due to MN as renal irAEs which is treatable with glucocorticoid.
Collapse
|
37
|
Lailler C, Lamuraglia M, Racine F, Louandre C, Godin C, Chauffert B, Galmiche A, Saidak Z. DNA damage response- and JAK-dependent regulation of PD-L1 expression in head and neck squamous cell carcinoma (HNSCC) cells exposed to 5-fluorouracil (5-FU). Transl Oncol 2021; 14:101110. [PMID: 33951601 PMCID: PMC8111315 DOI: 10.1016/j.tranon.2021.101110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 01/20/2023] Open
Abstract
PD-L1 is an important immune checkpoint molecule expressed by HNSCC. 5-FU induces PD-L1 expression in HNSCC cells. PD-L1 upregulation is DNA damage Response- and JAK-dependent. 5-FU potentiates the effect of the inflammatory cytokine Ifn-γ. Targeting EGFR with cetuximab blunts PD-L1 expression induced by 5-FU.
Objectives The immune checkpoint molecule PD-L1 (CD274) is a crucial regulator of the tumor immune response. Its expression has been reported in the therapeutic context in Head and Neck Squamous Cell Carcinoma (HNSCC), but it remains unclear how therapeutically approved molecules regulate PD-L1 expression in HNSCC cells. Materials and methods Three HNSCC cell lines (BICR6, PE/CA-PJ34 and PE/CA-PJ41) were used to analyze PD-L1 expression by immunoblotting, immunofluorescence and QPCR. Freely-available single cell RNAseq data from HNSCC were also used. Results 5-Fluorouracil (5-FU) increased the expression of PD-L1 with high efficacy in HNSCC cells. Single cell RNAseq data suggested the specificity of the regulation of PD-L1 in this context. The effect of 5-FU on PD-L1 expression was related to its genotoxic effect and was prevented by extracellular application of thymidine or using a chemical inhibitor of the DNA damage Response kinases ATM/ATR. We found that the effect of 5-FU was additive or synergistic with IFN-γ, the canonical inducer of PD-L1 in epithelial cells. QPCR analysis confirmed this finding and identified JAK-dependent transcriptional activation of PD-L1/CD274 as the underlying mechanism. The induction of PD-L1 by 5-FU was partially prevented by Epidermal Growth Factor Receptor (EGFR) inhibition with cetuximab. Conclusion Our study highlights the specific DNA Damage Response- and JAK- dependent induction of PD-L1 by 5-FU in HNSCC cells. This induction is regulated by the cytokine context and is potentially therapeutically actionable.
Collapse
Affiliation(s)
- Claire Lailler
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Sud, Amiens, France; UR7516 "CHIMERE", Université de Picardie Jules Verne, Amiens, France
| | - Michele Lamuraglia
- Laboratoire d'Imagerie Biomédicale (LIB), Sorbonne Université, CNRS, INSERM, Oncologie Médicale, CHU Sud, Amiens, France
| | - Floriane Racine
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Sud, Amiens, France; UR7516 "CHIMERE", Université de Picardie Jules Verne, Amiens, France
| | - Christophe Louandre
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Sud, Amiens, France; UR7516 "CHIMERE", Université de Picardie Jules Verne, Amiens, France
| | - Corinne Godin
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Sud, Amiens, France; UR7516 "CHIMERE", Université de Picardie Jules Verne, Amiens, France
| | - Bruno Chauffert
- UR7516 "CHIMERE", Université de Picardie Jules Verne, Amiens, France; Laboratoire d'Imagerie Biomédicale (LIB), Sorbonne Université, CNRS, INSERM, Oncologie Médicale, CHU Sud, Amiens, France
| | - Antoine Galmiche
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Sud, Amiens, France; UR7516 "CHIMERE", Université de Picardie Jules Verne, Amiens, France
| | - Zuzana Saidak
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Sud, Amiens, France; UR7516 "CHIMERE", Université de Picardie Jules Verne, Amiens, France.
| |
Collapse
|
38
|
Abstract
Trilaciclib (Cosela™) is a small-molecule, short-acting, inhibitor of cyclin-dependent kinases (CDK) 4 and 6 developed by G1 Therapeutics for its myeloprotection and potential antitumor efficacy and safety benefits in combination with cancer chemotherapy. CDKs govern cell cycle progression, and trilaciclib induces a transient, reversible G1 cell cycle arrest of proliferating haematopoietic stem and progenitor cells in bone marrow, thus protecting them from damage during chemotherapy. In February 2021, trilaciclib received its first approval in the USA to decrease the incidence of chemotherapy-induced myelosuppression in adult patients when administered prior to a platinum/etoposide-containing regimen or topotecan-containing regimen for extensive-stage small cell lung cancer (ES-SCLC). Clinical studies in breast cancer, colorectal cancer and small cell lung cancer are underway in several countries. This article summarizes the milestones in the development of trilaciclib leading to this first approval.
Collapse
Affiliation(s)
- Sohita Dhillon
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand.
| |
Collapse
|
39
|
Piecyk M, Triki M, Laval PA, Dragic H, Cussonneau L, Fauvre J, Duret C, Aznar N, Renno T, Manié SN, Chaveroux C, Ferraro-Peyret C. Pemetrexed Hinders Translation Inhibition upon Low Glucose in Non-Small Cell Lung Cancer Cells. Metabolites 2021; 11:metabo11040198. [PMID: 33810430 PMCID: PMC8067050 DOI: 10.3390/metabo11040198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/13/2021] [Accepted: 03/24/2021] [Indexed: 11/18/2022] Open
Abstract
Genetic alterations in non-small cell lung cancers (NSCLC) stimulate the generation of energy and biomass to promote tumor development. However, the efficacy of the translation process is finely regulated by stress sensors, themselves often controlled by nutrient availability and chemotoxic agents. Yet, the crosstalk between therapeutic treatment and glucose availability on cell mass generation remains understudied. Herein, we investigated the impact of pemetrexed (PEM) treatment, a first-line agent for NSCLC, on protein synthesis, depending on high or low glucose availability. PEM treatment drastically repressed cell mass and translation when glucose was abundant. Surprisingly, inhibition of protein synthesis caused by low glucose levels was partially dampened upon co-treatment with PEM. Moreover, PEM counteracted the elevation of the endoplasmic reticulum stress (ERS) signal produced upon low glucose availability, providing a molecular explanation for the differential impact of the drug on translation according to glucose levels. Collectively, these data indicate that the ERS constitutes a molecular crosstalk between microenvironmental stressors, contributing to translation reprogramming and proteostasis plasticity.
Collapse
Affiliation(s)
- Marie Piecyk
- Cancer Research Centre of Lyon, Université Lyon, INSERM 1052, CNRS 5286, 69008 Lyon, France; (M.P.); (M.T.); (P.-A.L.); (H.D.); (J.F.); (C.D.); (N.A.); (T.R.)
- Hospices Civils de Lyon, Biopathology of Tumours, CPE, GHE Hospital, 69500 Bron, France
| | - Mouna Triki
- Cancer Research Centre of Lyon, Université Lyon, INSERM 1052, CNRS 5286, 69008 Lyon, France; (M.P.); (M.T.); (P.-A.L.); (H.D.); (J.F.); (C.D.); (N.A.); (T.R.)
| | - Pierre-Alexandre Laval
- Cancer Research Centre of Lyon, Université Lyon, INSERM 1052, CNRS 5286, 69008 Lyon, France; (M.P.); (M.T.); (P.-A.L.); (H.D.); (J.F.); (C.D.); (N.A.); (T.R.)
| | - Helena Dragic
- Cancer Research Centre of Lyon, Université Lyon, INSERM 1052, CNRS 5286, 69008 Lyon, France; (M.P.); (M.T.); (P.-A.L.); (H.D.); (J.F.); (C.D.); (N.A.); (T.R.)
| | - Laura Cussonneau
- INRAE, Unité de Nutrition Humaine, Université Clermont Auvergne, UMR1019, 63122 Clermont-Ferrand, France;
| | - Joelle Fauvre
- Cancer Research Centre of Lyon, Université Lyon, INSERM 1052, CNRS 5286, 69008 Lyon, France; (M.P.); (M.T.); (P.-A.L.); (H.D.); (J.F.); (C.D.); (N.A.); (T.R.)
| | - Cédric Duret
- Cancer Research Centre of Lyon, Université Lyon, INSERM 1052, CNRS 5286, 69008 Lyon, France; (M.P.); (M.T.); (P.-A.L.); (H.D.); (J.F.); (C.D.); (N.A.); (T.R.)
| | - Nicolas Aznar
- Cancer Research Centre of Lyon, Université Lyon, INSERM 1052, CNRS 5286, 69008 Lyon, France; (M.P.); (M.T.); (P.-A.L.); (H.D.); (J.F.); (C.D.); (N.A.); (T.R.)
| | - Toufic Renno
- Cancer Research Centre of Lyon, Université Lyon, INSERM 1052, CNRS 5286, 69008 Lyon, France; (M.P.); (M.T.); (P.-A.L.); (H.D.); (J.F.); (C.D.); (N.A.); (T.R.)
| | - Serge N. Manié
- Inserm U1242, Centre de Lutte Contre le Cancer Eugène Marquis, Université de Rennes, 35042 Rennes, France;
| | - Cédric Chaveroux
- Cancer Research Centre of Lyon, Université Lyon, INSERM 1052, CNRS 5286, 69008 Lyon, France; (M.P.); (M.T.); (P.-A.L.); (H.D.); (J.F.); (C.D.); (N.A.); (T.R.)
- Correspondence: (C.C.); (C.F.-P.)
| | - Carole Ferraro-Peyret
- Cancer Research Centre of Lyon, Université Lyon, INSERM 1052, CNRS 5286, 69008 Lyon, France; (M.P.); (M.T.); (P.-A.L.); (H.D.); (J.F.); (C.D.); (N.A.); (T.R.)
- Hospices Civils de Lyon, Biopathology of Tumours, CPE, GHE Hospital, 69500 Bron, France
- Correspondence: (C.C.); (C.F.-P.)
| |
Collapse
|