1
|
Hu Q, Huang KC, Nakajo K, Zhang Y, Qiu H. Initial Treatment Modalities in Patients with Newly Diagnosed Primary Lung Cancer in Japan. Curr Oncol 2025; 32:32. [PMID: 39851948 PMCID: PMC11763771 DOI: 10.3390/curroncol32010032] [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: 11/25/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
Treatment for lung cancer continues to rapidly evolve. Here, we describe trends in the initial treatment of adults with newly diagnosed primary non-small-cell lung cancer in Japan. This retrospective cohort study used data from JMDC Inc. Claims Database from 2015 to 2023. Adults with lung cancer, confirmed using a combination of diagnosis, treatment, or procedure codes, were enrolled. A total of 9373 patients were included, with a mean age of approximately 59 years. The median time from diagnosis to treatment initiation ranged from 38 days in patients treated surgically to 25 days in patients with distant metastases. The observed trends were a decrease in the percentage of newly diagnosed patients with distant metastases, a decline in chemotherapy use in patients with early-stage disease, and in advanced disease, a more than doubling in the use of targeted therapy, including checkpoint inhibitors, while radiotherapy and chemotherapy tended to decrease. The observed changes in treatment were driven mainly by the increased use of targeted therapies including checkpoint inhibitors and are aligned with current treatment guidelines in Japan. The observation of fewer patients with distant metastases over time possibly indicates earlier detection. Additional research is needed to understand if new therapies are being extended to older and frail patients with lung cancer in Japan.
Collapse
Affiliation(s)
- Qingqing Hu
- Global Epidemiology, Office of the Chief Medical Officer, Janssen Research & Development, LLC, Shanghai 200231, China; (Q.H.); (Y.Z.)
| | - Kuan-Chih Huang
- Global Epidemiology, Office of the Chief Medical Officer, Janssen Research & Development, LLC, Taipei 104, Taiwan;
| | - Ko Nakajo
- Global Epidemiology, Office of the Chief Medical Officer, Janssen Research & Development, LLC, Tokyo 101-0065, Japan;
| | - Yongjing Zhang
- Global Epidemiology, Office of the Chief Medical Officer, Janssen Research & Development, LLC, Shanghai 200231, China; (Q.H.); (Y.Z.)
| | - Hong Qiu
- Global Epidemiology, Office of the Chief Medical Officer, Janssen Research & Development, LLC, Titusville, NJ 08560, USA
| |
Collapse
|
2
|
Yu KX, Yuan WJ, Jing-Li, Wang HZ, Li YX. A Comprehensive Pan-cancer Analysis Identified that TRIB3 was Associated with Immune Cell Infiltration and Poor Prognosis. Curr Pharm Biotechnol 2025; 26:878-901. [PMID: 39279106 DOI: 10.2174/0113892010308103240830063504] [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: 03/13/2024] [Revised: 06/28/2024] [Accepted: 07/19/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Previous studies have demonstrated that TRIB3 plays a carcinogenic role in tumor progression. However, the exploration of TRIB3 at the pan-cancer level has not been reported. AIMS This study aimed to conduct a comprehensive pan-cancer analysis of TRIB3. OBJECTIVES We explored the expression pattern and functional mechanism of TRIB3 on the basis of multiple databases. METHODS We first explored the expression level of TRIB3 in the TCGA database. Then, the receiver operation characteristic curve (ROC), Kaplan-Meier plotter, and Cox regression were used to estimate the diagnostic and prognostic value of TRIB3, respectively. We also explored the relationship between TRIB3 and the infiltration of tumor immune cells, as well as the expression of immune checkpoint molecules. Gene enrichment and protein interaction network analysis were carried out to identify possible carcinogenic molecular mechanisms and functional pathways. Finally, we compared the non-promoter region methylation of TRIB3 in normal and tumor tissues and explored potential systems with unique functions in TRIB3-mediated tumorigenesis. RESULTS The expression level of TRIB3 was elevated in multiple tumor types, and the high expression of TRIB3 was associated with poor prognosis. TRIB3 had a higher frequency of genetic changes in several tumors and showed varying trends in TRIB3 methylation levels. Additionally, high expression of TRIB3 was also associated with infiltration of cancer-related fibroblasts and different types of immune cells and was positively correlated with the expression of immune checkpoint molecules. Furthermore, gene enrichment analysis suggested that TRIB3 may play a role in the malignant progression of cancer by participating in protein post-translational modifications and activating transcription initiation factors. CONCLUSION Our pan-cancer analysis provided the potential carcinogenic role of TRIB3 in tumors and verified a promising target for clinical immune treatment.
Collapse
Affiliation(s)
- Ke-Xun Yu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei-Jie Yuan
- Department of Gastrointestinal Surgery, Xiangya Hospital of Central South University, Changsha, China
| | - Jing-Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui-Zhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yong-Xiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Saha S, Ghosh S, Ghosh S, Nandi S, Nayak A. Unraveling the complexities of colorectal cancer and its promising therapies - An updated review. Int Immunopharmacol 2024; 143:113325. [PMID: 39405944 DOI: 10.1016/j.intimp.2024.113325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Colorectal cancer (CRC) continues to be a global health concern, necessitating further research into its complex biology and innovative treatment approaches. The etiology, pathogenesis, diagnosis, and treatment of colorectal cancer are summarized in this thorough review along with recent developments. The multifactorial nature of colorectal cancer is examined, including genetic predispositions, environmental factors, and lifestyle decisions. The focus is on deciphering the complex interactions between signaling pathways such as Wnt/β-catenin, MAPK, TGF-β as well as PI3K/AKT that participate in the onset, growth, and metastasis of CRC. There is a discussion of various diagnostic modalities that span from traditional colonoscopy to sophisticated molecular techniques like liquid biopsy and radiomics, emphasizing their functions in early identification, prognostication, and treatment stratification. The potential of artificial intelligence as well as machine learning algorithms in improving accuracy as well as efficiency in colorectal cancer diagnosis and management is also explored. Regarding therapy, the review provides a thorough overview of well-known treatments like radiation, chemotherapy, and surgery as well as delves into the newly-emerging areas of targeted therapies as well as immunotherapies. Immune checkpoint inhibitors as well as other molecularly targeted treatments, such as anti-epidermal growth factor receptor (anti-EGFR) as well as anti-vascular endothelial growth factor (anti-VEGF) monoclonal antibodies, show promise in improving the prognosis of colorectal cancer patients, in particular, those suffering from metastatic disease. This review focuses on giving readers a thorough understanding of colorectal cancer by considering its complexities, the present status of treatment, and potential future paths for therapeutic interventions. Through unraveling the intricate web of this disease, we can develop a more tailored and effective approach to treating CRC.
Collapse
Affiliation(s)
- Sayan Saha
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Shreya Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Suman Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Sumit Nandi
- Department of Pharmacology, Gupta College of Technological Sciences, Asansol, West Bengal 713301, India
| | - Aditi Nayak
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India.
| |
Collapse
|
4
|
Zhang G, Liu J, Li S, Wang T, Chen L, Li H, Ding Q, Li X, Zhu S, Tang X. Cytochalasin H enhances sensitivity to gefitinib in non-small-cell lung cancer cells through inhibiting EGFR activation and PD-L1 expression. Sci Rep 2024; 14:25276. [PMID: 39455693 PMCID: PMC11512071 DOI: 10.1038/s41598-024-76060-2] [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: 06/14/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
In our previous study, we have isolated cytochalasin H (CyH) from endophytic fungus derived from mangrove plant and found that CyH inhibited the proliferation of non-small cell lung cancer (NSCLC) cells. Recently, epidermal growth factor receptor (EGFR) activation and programmed cell death 1 ligand (PD-L1) expression have been demonstrated to mediate NSCLC resistance to gefitinib, first-generation EGFR tyrosine kinase inhibitor (EGFR-TKI). Here, we further investigated the effect of CyH on EGFR activation, PD-L1 expression, and gefitinib sensitivity in NSCLC cell lines, A549 (wild-type EGFR), HCC827 (EGFR mutation), and NCI-H1975 (dual EGFR mutations and acquired gefitinib resistance) and animal model. Our results showed that CyH significantly inhibited EGFR activation and PD-L1 expression in NSCLC cells. Additionally, CyH dramatically promoted the inhibitory effect of gefitinib on the proliferation of A549 and HCC827 cells, and enhanced the sensitivity to gefitinib in NCI-H1975 cells. Moreover, CyH increased the inhibitory effect of gefitinib on EGFR activation and PD-L1 expression in HCC827 and NCI-H1975 cells. Animal experiments further demonstrated that CyH significantly promoted the inhibitory effect of gefitinib on the growth of NSCLC and the expression of Ki-67, p-EGFR, and PD-L1 in NCI-H1975 NSCLC xenograft tumors of nude mice. Furthermore, CyH inhibited the activation of JAK3/STAT signaling pathway. Taken together, our findings suggest that CyH promotes the sensitivity to gefitinib in NSCLC cells through the inhibition of EGFR activation and PD-L1 expression.
Collapse
Affiliation(s)
- Guihong Zhang
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Jiao Liu
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Sanzhong Li
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Tianyu Wang
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Li Chen
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Huan Li
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Qingkai Ding
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Xiangyong Li
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan, 523808, People's Republic of China
| | - Shaoping Zhu
- Center for Animal, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Xudong Tang
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China.
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan, 523808, People's Republic of China.
| |
Collapse
|
5
|
Penkov K, Bondarenko I, Saenko DV, Kulyaba Y, Guo J, Gong Y, Yamamoto N, Hotko YS, Boyko V, Fadeeva NV, Ursol GM, Ahn HK, Kislov NV, Shen CI, Davis C, Kowalski K, Michelon E, Pavlov D, Hirohashi T, Cho BC. Pharmacokinetics, safety, and efficacy of an alternative dosing regimen of sasanlimab in participants with advanced NSCLC and other malignancies. Ther Adv Med Oncol 2024; 16:17588359241274592. [PMID: 39281971 PMCID: PMC11393800 DOI: 10.1177/17588359241274592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/02/2024] [Indexed: 09/18/2024] Open
Abstract
Background Sasanlimab (PF-06801591), a humanized immunoglobulin G4 monoclonal antibody, binds to programmed cell death protein-1 (PD-1), preventing ligand (PD-L1) interaction. Objectives To evaluate pharmacokinetics (PK), safety, tolerability, and efficacy of two subcutaneous sasanlimab dosing regimens. Design An open-label study consisting of phases Ib and II. Phase Ib: non-randomized, dose escalation, and expansion study in Asian participants with advanced malignancies. Phase II conducted globally in participants with non-small-cell lung cancer with PD-L1 positive or PD-L1 status unknown tumors; participants were randomized 1:2 to receive subcutaneous sasanlimab 300 mg once every 4 weeks (300 mg-Q4W) or 600 mg once every 6 weeks (600 mg-Q6W). Methods Primary endpoint in phase Ib: dose-limiting toxicity (DLT) occurring in first treatment cycle; in phase II: C trough and AUC. Results A total of 155 participants (phase Ib, n = 34; phase II, n = 121) received sasanlimab. Phase Ib: no DLT reported. Phase II: ratio of adjusted geometric mean for AUCtau was 231.2 (90% CI, 190.1-281.2) and C trough was 111.5 (90% CI, 86.3-144.0) following 600 mg-Q6W (test) versus 300 mg-Q4W (reference). Phase Ib: grade 3 treatment-related adverse events (TRAEs) occurred in 1/4 (25%) and 3/12 (25%) participants treated in 300 mg-Q4W dose escalation and expansion cohorts, respectively. Phase II: grade 3 TRAEs occurred in 3/41 (7.3%) and 3/80 (3.8%) participants treated with 300 mg-Q4W and 600 mg-Q6W, respectively; no grade 4/5 TRAEs. Phase II: confirmed objective response was observed in 11/41 (26.8% (95% CI, 14.2-42.9)) and 12/80 (15.0% (95% CI, 8.0-24.7)) participants treated with 300 mg-Q4W and 600 mg-Q6W, respectively. Conclusions Phase Ib regimens were considered safe with no DLTs reported. In phase II, 600 mg-Q6W regimen criteria were met for AUCtau and C trough metrics to support PK-based extrapolation of efficacy of alternative regimen. Regimens were well tolerated, showing anti-tumor activity in participants with advanced solid tumors. Administration of sasanlimab at a dose of 600 mg-Q6W subcutaneously may serve as a convenient alternative to 300 mg-Q4W administration. Trial registration NCT04181788 (ClinicalTrials.gov); 2019-003818-14 (EudraCT).
Collapse
Affiliation(s)
- Konstantin Penkov
- Private Medical Institution "Euromedservice," Saint-Petersburg, Russia
| | | | | | | | - Jun Guo
- Beijing Cancer Hospital, Beijing, China
| | - Yi Gong
- Chongqing University Cancer Hospital, Chongqing, China
| | | | - Yevhen Stepanovych Hotko
- Municipal nonprofit enterprise Central City Clinical Hospital of Uzhhorod City Council, Uzhgorod, Ukraine
| | - Vasyl Boyko
- Municipal Non-profit Enterprise "SubCarpathian Clinical Oncological Centre of Ivano-Frankivsk RC," Ivano-Frankivsk, Ukraine
| | | | | | - Hee Kyung Ahn
- Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Nikolay Viktorovich Kislov
- State Budgetary Institution of Healthcare of Yaroslavl Region "Clinical Oncology Hospital," Yaroslavl, Russia
| | - Chia-I Shen
- Taipei Veterans General Hospital, Taipei, Taiwan
| | | | | | | | | | | | - Byoung Chul Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
6
|
Bai L, Liu H, You R, Jiang X, Zhang T, Li Y, Shan T, Qian Z, Wang Y, Liu Y, Li C. Combination Nano-Delivery Systems Remodel the Immunosuppressive Tumor Microenvironment for Metastatic Triple-Negative Breast Cancer Therapy. Mol Pharm 2024; 21:2148-2162. [PMID: 38536949 DOI: 10.1021/acs.molpharmaceut.3c00242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer for which effective therapies are lacking. Targeted remodeling of the immunosuppressive tumor microenvironment (TME) and activation of the body's immune system to fight tumors with well-designed nanoparticles have emerged as pivotal breakthroughs in tumor treatment. To simultaneously remodel the immunosuppressive TME and trigger immune responses, we designed two potential therapeutic nanodelivery systems to inhibit TNBC. First, the bromodomain-containing protein 4 (BRD4) inhibitor JQ1 and the cyclooxygenase-2 (COX-2) inhibitor celecoxib (CXB) were coloaded into chondroitin sulfate (CS) to obtain CS@JQ1/CXB nanoparticles (NPs). Then, the biomimetic nanosystem MM@P3 was prepared by coating branched polymer poly(β-amino ester) self-assembled NPs with melittin embedded macrophage membranes (MM). Both in vitro and in vivo, the CS@JQ1/CXB and MM@P3 NPs showed excellent immune activation efficiencies. Combination treatment exhibited synergistic cytotoxicity, antimigration ability, and apoptosis-inducing and immune activation effects on TNBC cells and effectively suppressed tumor growth and metastasis in TNBC tumor-bearing mice by activating the tumor immune response and inhibiting angiogenesis. In summary, this study offers a novel combinatorial immunotherapeutic strategy for the clinical TNBC treatment.
Collapse
Affiliation(s)
- Liya Bai
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin 300070, China
| | - Hui Liu
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin 300070, China
| | - Ran You
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xiaoyu Jiang
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin 300070, China
| | - Tao Zhang
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin 300070, China
| | - Yunan Li
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin 300070, China
| | - Tianhe Shan
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin 300070, China
| | - Zhanyin Qian
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin 300070, China
| | - Yinsong Wang
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yuanyuan Liu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Chunyu Li
- Department of Integrated Traditional Chinese and Western Medicine, International Medical School, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
7
|
Chen X, Ye L, Wang H, Liu X, Zhao L, Xu K, Liu Y, He Y. Promising preclinical models for lung cancer research-lung cancer organoids: a narrative review. Transl Lung Cancer Res 2024; 13:623-634. [PMID: 38601435 PMCID: PMC11002517 DOI: 10.21037/tlcr-23-341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 01/17/2024] [Indexed: 04/12/2024]
Abstract
Background and Objective Traditional cell line models are the commonly used preclinical models for lung cancer research. However, cell lines cannot recapitulate the complex tumor heterogeneity and cannot mimic the microenvironment of human cancer. Recently, 3D multicellular in vitro self-assembled models called "organoids" have been developed at a fast pace in the field of research, which can mimic the actual primary tumor. At present, several studies have reported on protocols of lung cancer organoids (LCOs) generation, and using LCOs can provide novel insight into the basic and translational research of lung cancer. However, the establishment of the LCO models remains challenging due to the complexity of lung cancer and the immaturity of organoid technology, so it is necessary to understand the influences of different methodologies on LCO generation and review the applications and limitations of LCO models. Methods In this review, we searched the literature in the recent ten years in the field of LCOs. Key Content and Findings We summarized the methodology, the problems, and the solutions in the LCOs generation, its application and limitations, as well as proposing future challenges and perspectives. Conclusions Currently, LCOs are successfully generated via exploring the methodology by the researchers. Though there are still challenges in clinical application, LCOs are applied in some cancer studies including investigation of anti-cancer treatment response in vitro, modeling tumor immune microenvironment, and construction of organ chips, which are forging a promising path towards precision medicine.
Collapse
Affiliation(s)
- Xinru Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Li Ye
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Xinyue Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Lishu Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Kandi Xu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Yujin Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
8
|
Barr T, Ma S, Li Z, Yu J. Recent advances and remaining challenges in lung cancer therapy. Chin Med J (Engl) 2024; 137:533-546. [PMID: 38321811 DOI: 10.1097/cm9.0000000000002991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Indexed: 02/08/2024] Open
Abstract
ABSTRACT Lung cancer remains the most common cause of cancer death. Given the continued research into new drugs and combination therapies, outcomes in lung cancer have been improved, and clinical benefits have been expanded to a broader patient population. However, the overall cure and survival rates for lung cancer patients remain low, especially in metastatic cases. Among the available lung cancer treatment options, such as surgery, radiation therapy, chemotherapy, targeted therapies, and alternative therapies, immunotherapy has shown to be the most promising. The exponential progress in immuno-oncology research and recent advancements made in the field of immunotherapy will further increase the survival and quality of life for lung cancer patients. Substantial progress has been made in targeted therapies using tyrosine kinase inhibitors and monoclonal antibody immune checkpoint inhibitors with many US Food And Drug Administration (FDA)-approved drugs targeting the programmed cell death ligand-1 protein (e.g., durvalumab, atezolizumab), the programmed cell death-1 receptor (e.g., nivolumab, pembrolizumab), and cytotoxic T-lymphocyte-associated antigen 4 (e.g., tremelimumab, ipilimumab). Cytokines, cancer vaccines, adoptive T cell therapies, and Natural killer cell mono- and combinational therapies are rapidly being studied, yet to date, there are currently none that are FDA-approved for the treatment of lung cancer. In this review, we discuss the current lung cancer therapies with an emphasis on immunotherapy, including the challenges for future research and clinical applications.
Collapse
Affiliation(s)
- Tasha Barr
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California 91010, USA
| | - Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California 91010, USA
- Comprehensive Cancer Center, City of Hope, Los Angeles, California 91010, USA
| | - Zhixin Li
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California 91010, USA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California 91010, USA
- Comprehensive Cancer Center, City of Hope, Los Angeles, California 91010, USA
- Department of Immuno-Oncology, Beckman Research Institute, Los Angeles, California 91010, USA
| |
Collapse
|
9
|
Wang SH, Cao Z, Farazuddin M, Chen J, Janczak KW, Tang S, Cannon J, Baker JR. A novel intranasal peptide vaccine inhibits non-small cell lung cancer with KRAS mutation. Cancer Gene Ther 2024; 31:464-471. [PMID: 38177307 DOI: 10.1038/s41417-023-00717-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024]
Abstract
KRAS mutations occur commonly in the lung and can lead to the development of non-small cell lung cancer (NSCLC). While the mutated KRAS protein is a neoantigen, it usually does not generate an effective anti-tumor immune response on mucosal/epithelial surfaces. Despite this, mutated KRAS remains a potential target for immunotherapy since immune targeting of this protein in animal models has been effective at eliminating tumor cells. We attempted to develop a KRAS vaccine using mutated and wild-type KRAS peptides in combination with a nanoemulsion (NE) adjuvant. The efficacy of this approach was tested in an inducible mutant KRAS-mouse lung tumor model. Animals were immunized intranasally using NE with KRAS peptides. These animals had decreased CD4+FoxP3+ T cells in both lymph nodes and spleen. Immunized animals also showed higher IFN-γ and IL-17a levels to mutated KRAS that were produced by CD8+ T cells and enhancement in KRAS-specific Th1 and Th17 responses that persisted for 3 months after the last vaccination. Importantly, the immunized animals had significantly decreased tumor incidence compared to control animals. In conclusion, a mucosal approach to KRAS vaccination demonstrated the ability to induce local KRAS-specific immune responses in the lung and resulted in reduced tumor incidence.
Collapse
Affiliation(s)
- Su He Wang
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Medical School, University of Michigan, Ann Arbor, MI, 48109, USA.
- Division of Allergy, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Zhengyi Cao
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Medical School, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mohammad Farazuddin
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Medical School, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Allergy, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jesse Chen
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Medical School, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Katarzyna W Janczak
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Medical School, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shengzhuang Tang
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Medical School, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jayme Cannon
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Medical School, University of Michigan, Ann Arbor, MI, 48109, USA
| | - James R Baker
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Medical School, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Allergy, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
10
|
Pan L, Hu L, Chen M, Song Y, Chen Z, Gu Y, Li C, Jiang Z. A novel CD47-blocking peptide fused to pro-apoptotic KLA repeat inhibits lung cancer growth in mice. Cancer Immunol Immunother 2023; 72:4179-4194. [PMID: 37831145 PMCID: PMC10992817 DOI: 10.1007/s00262-023-03554-9] [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: 06/08/2023] [Accepted: 09/22/2023] [Indexed: 10/14/2023]
Abstract
CD47 is highly expressed in many tumor tissues and induces immune evasion by interaction with SIRP-alpha (signal regulatory protein-alpha) expressed on tumor-associated macrophages. In this study, we identified a novel CD47-blocking peptide VK17 by phage display technique. A pro-apoptotic VK30 peptide was obtained after VK17 was fused to KLA amino acid repeat at C-termini. The VK30 was specifically bound to CD47 on lung cancer cells, and subsequently inducing lung cancer cell apoptosis. Meanwhile, the expression of Bax was increased, whereas the expression of Bcl-2 and Ki-67 were reduced in the VK30-treated lung cancer cells. In addition, VK30 effectively improved the phagocytic activity of macrophages against VK30-pretreated lung cancer cells. Combinational treatment of lung cancer cells with blocking antibody anti-CD47 and VK30 additively enhanced VK30 binding to CD47, subsequently increasing lung cancer cell apoptosis and macrophage phagocytosis. Intraperitoneal administration of 2 mg/kg VK30 induced effective trafficking of VK30 into tumor tissues, and suppressing lung cancer cell growth in mice, associated with increased tumor cell apoptosis, macrophage activation and phagocytosis in vivo. The expression of CD47 was reduced in the VK30-treated tumor tissues and the expression level was positively correlated to tumor size. In addition, VK30 reduced the infiltration of CD11b+Ly6G+ neutrophils and CD11b+Ly6C+Ly6G+ granulocytic myeloid-derived suppressor cells (Gr-MDSCs) in tumor tissues, associated with suppressed expression of tumorigenic IL-6 and TNF-alpha from these cell types. Thereby, VK30 exerted anti-tumor effects in mice through inducing tumor cell apoptosis and macrophage phagocytosis. VK30 would be a novel therapeutic peptide in lung cancer immunotherapy.
Collapse
Affiliation(s)
- Linyue Pan
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Lu Hu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengjie Chen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
| | - Zhihong Chen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yutong Gu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chun Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Zhilong Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
11
|
Kuah CY, Monfries R, Quartagno M, Seckl MJ, Ghorani E. What is the optimal duration, dose and frequency for anti-PD1 therapy of non-small cell lung cancer? Ther Adv Med Oncol 2023; 15:17588359231210271. [PMID: 37954230 PMCID: PMC10638879 DOI: 10.1177/17588359231210271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Over the past decade, immune checkpoint inhibitors (ICIs) have transformed the management of multiple malignancies including lung cancer. However, the optimal use of these agents in terms of duration, dose and administration frequency remains unknown. Focusing on anti-PD1 agents nivolumab and pembrolizumab in the context of non-small cell lung cancer, we argue that several lines of evidence suggest current administration regimens of these drugs may result in overtreatment with potentially important implications for cost, quality of life and toxicity. This review summarizes evidence for the scope to optimize anti-PD1 regimens, the limitations of existing data and potential approaches to solve these problems including with a novel multi-arm clinical trial design implemented in the recently opened REFINE-Lung study.
Collapse
Affiliation(s)
- Chii Yang Kuah
- Department of Medical Oncology, Charing Cross Hospital Campus of Imperial College London, UK
| | - Robert Monfries
- Department of Medical Oncology, Charing Cross Hospital Campus of Imperial College London, UK
| | - Matteo Quartagno
- Institute for Clinical Trials and Methodology, University College London, London, UK
| | - Michael J. Seckl
- Department of Medical Oncology, Charing Cross Hospital Campus of Imperial College London W6 8RF, UK
| | - Ehsan Ghorani
- Department of Medical Oncology, Charing Cross Hospital Campus of Imperial College London W6 8RF, UK
| |
Collapse
|
12
|
Zuo B, Li T, Liu X, Wang S, Cheng J, Liu X, Cui W, Shi H, Ling C. Dipeptidyl peptidase 4 inhibitor reduces tumor-associated macrophages and enhances anti-PD-L1-mediated tumor suppression in non-small cell lung cancer. Clin Transl Oncol 2023; 25:3188-3202. [PMID: 37115489 PMCID: PMC10514125 DOI: 10.1007/s12094-023-03187-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/03/2023] [Indexed: 04/29/2023]
Abstract
PURPOSE The efficacy of immune checkpoint inhibitors such as programmed cell death ligand 1 (PD-L1) antibodies in non-small cell lung cancer (NSCLC) is limited, and combined use with other therapies is recommended. Dipeptidyl peptidase 4 (DPP4) inhibitors, a class of small molecule inhibitors, are highly effective for treating type 2 diabetes. Emerging evidence implicates DPP4 inhibitors as immunomodulators that modify aspects of innate and adaptive immunity. We evaluated the combination of a DPP4 inhibitor (anagliptin) and PD-L1 blockade in an NSCLC mouse model. METHODS The effect of the combination of anti-PD-L1 and anagliptin was evaluated in subcutaneous mouse models of NSCLC. Tumor-infiltrating immune cells were analyzed by flow cytometry. Bone marrow-derived monocytes of C57BL/6 mice were isolated in vitro to examine the underlying mechanism of anagliptin on the differentiation and polarization of macrophage. RESULTS Anagliptin dramatically improved the efficacy of PD-L1 antibody monotherapy by inhibiting macrophage formation and M2 polarization in the tumor microenvironment. Mechanistically, anagliptin suppressed the production of reactive oxygen species in bone marrow monocytes by inhibiting NOX1 and NOX2 expression induced by macrophage colony-stimulating factor, reduced late ERK signaling pathway activation, and inhibited monocyte-macrophage differentiation. However, the inhibitory effect was reactivated by lipopolysaccharide and interferon-gamma interacting with corresponding receptors during M1 macrophage polarization, but not M2. CONCLUSIONS Anagliptin can enhance PD-L1 blockade efficacy in NSCLC by inhibiting macrophage differentiation and M2 macrophage polarization, and combination therapy may be a promising strategy for treating PD-L1 blockade therapy-resistant patients with NSCLC.
Collapse
Affiliation(s)
- Bei Zuo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, China
- Department of Respiratory and Critical Care Medicine, The Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221116, China
| | - Tao Li
- Department of Respiratory and Critical Care Medicine, The Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221116, China
| | - Xiaoyun Liu
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Shuling Wang
- Department of Respiratory and Critical Care Medicine, The Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221116, China
| | - Jianxiang Cheng
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| | - Xiangqun Liu
- Department of Respiratory and Critical Care Medicine, The Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221116, China
| | - Wenjie Cui
- Department of Respiratory and Critical Care Medicine, The Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221116, China
| | - Hengliang Shi
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, China.
| | - Chunhua Ling
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China.
| |
Collapse
|
13
|
Castillo DR, Jeon WJ, Park D, Pham B, Yang C, Joung B, Moon JH, Lee J, Chong EG, Park K, Reeves ME, Duerksen-Hughes P, Mirshahidi HR, Mirshahidi S. Comprehensive Review: Unveiling the Pro-Oncogenic Roles of IL-1ß and PD-1/PD-L1 in NSCLC Development and Targeting Their Pathways for Clinical Management. Int J Mol Sci 2023; 24:11547. [PMID: 37511306 PMCID: PMC10380530 DOI: 10.3390/ijms241411547] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
In the past decade, targeted therapies for solid tumors, including non-small cell lung cancer (NSCLC), have advanced significantly, offering tailored treatment options for patients. However, individuals without targetable mutations pose a clinical challenge, as they may not respond to standard treatments like immune-checkpoint inhibitors (ICIs) and novel targeted therapies. While the mechanism of action of ICIs seems promising, the lack of a robust response limits their widespread use. Although the expression levels of programmed death ligand 1 (PD-L1) on tumor cells are used to predict ICI response, identifying new biomarkers, particularly those associated with the tumor microenvironment (TME), is crucial to address this unmet need. Recently, inflammatory cytokines such as interleukin-1 beta (IL-1β) have emerged as a key area of focus and hold significant potential implications for future clinical practice. Combinatorial approaches of IL-1β inhibitors and ICIs may provide a potential therapeutic modality for NSCLC patients without targetable mutations. Recent advancements in our understanding of the intricate relationship between inflammation and oncogenesis, particularly involving the IL-1β/PD-1/PD-L1 pathway, have shed light on their application in lung cancer development and clinical outcomes of patients. Targeting these pathways in cancers like NSCLC holds immense potential to revolutionize cancer treatment, particularly for patients lacking targetable genetic mutations. However, despite these promising prospects, there remain certain aspects of this pathway that require further investigation, particularly regarding treatment resistance. Therefore, the objective of this review is to delve into the role of IL-1β in NSCLC, its participation in inflammatory pathways, and its intricate crosstalk with the PD-1/PD-L1 pathway. Additionally, we aim to explore the potential of IL-1β as a therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Dani Ran Castillo
- Division of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA; (D.R.C.); (E.G.C.); (M.E.R.); (H.R.M.)
| | - Won Jin Jeon
- Department of Internal Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.J.J.); (B.P.); (B.J.); (J.H.M.)
| | - Daniel Park
- Department of Internal Medicine, University of San Francisco-Fresno, Fresno, CA 93701, USA;
| | - Bryan Pham
- Department of Internal Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.J.J.); (B.P.); (B.J.); (J.H.M.)
| | - Chieh Yang
- Department of Internal Medicine, School of Medicine, University of California Riverside, Riverside, CA 92521, USA;
| | - Bowon Joung
- Department of Internal Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.J.J.); (B.P.); (B.J.); (J.H.M.)
| | - Jin Hyun Moon
- Department of Internal Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.J.J.); (B.P.); (B.J.); (J.H.M.)
| | - Jae Lee
- School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Esther G. Chong
- Division of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA; (D.R.C.); (E.G.C.); (M.E.R.); (H.R.M.)
| | - Kiwon Park
- Department of Pharmacy, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Mark E. Reeves
- Division of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA; (D.R.C.); (E.G.C.); (M.E.R.); (H.R.M.)
| | - Penelope Duerksen-Hughes
- Division of Biochemistry, Department of Medicine & Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Hamid R. Mirshahidi
- Division of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA; (D.R.C.); (E.G.C.); (M.E.R.); (H.R.M.)
| | - Saied Mirshahidi
- Biospecimen Laboratory, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA
- Division of Microbiology and Molecular Genetics, Department of Medicine & Basic Sciences, Loma Linda University, Loma Linda 92350, CA, USA
| |
Collapse
|
14
|
Luo P, Li S, Long X. N6-methyladenosine RNA modification in PD-1/PD-L1: Novel implications for immunotherapy. Biochim Biophys Acta Rev Cancer 2023; 1878:188873. [PMID: 36842764 DOI: 10.1016/j.bbcan.2023.188873] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 02/26/2023]
Abstract
Cancer immunotherapy has been shown to achieve significant antitumor effects in a variety of malignancies. Out of all the immune checkpoint molecules, PD-1/PD-L1 inhibitor therapy has achieved great success. However, only some cancer patients benefit from this treatment strategy owing to drug resistance. Therefore, identifying the underlying modulators of the PD-1/PD-L1 pathway to completely comprehend the mechanisms of anti-PD-1/PD-L1 treatment is crucially important. Recent research has validated that m6A modification plays a critical role in the PD-1/PD-L1 axis, thus regulating the immune response and immunotherapy strategies. In this review, we summarized the latest research on the regulation of m6A modification in PD-1/PD-L1 pathways in cancer proliferation, invasion, and prognosis based on different kinds of cancers and discussed the possible mechanisms. We also reviewed m6A-associated lncRNAs in the regulation of the PD-1/PD-L1 pathway. More importantly, we outlined the influence of m6A modulation on anti-PD-1 therapy and m6A-related molecules that could predict the curative effect of anti-PD-1/PD-L1 therapy. Further studies exploring the definitive regulation of m6A on the PD1/PD-1 pathway and immunotherapy are needed, which may address some of the current limitations in immunotherapy.
Collapse
Affiliation(s)
- Ping Luo
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shiqi Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xinghua Long
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|