1
|
Luo J, Li L, Wang H, Zhang X, He F, Shi M, Zhang X, Tang R, Bao Y. Analysis of therapeutic effects and influencing factors of ICIs in lung-cancer patients. Clin Transl Oncol 2025; 27:2597-2604. [PMID: 39560833 DOI: 10.1007/s12094-024-03767-z] [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: 08/10/2024] [Accepted: 10/13/2024] [Indexed: 11/20/2024]
Abstract
OBJECTIVE The aim of this retrospective study was to analyze the efficacy and risk factors of immune checkpoint inhibitors (ICIs) in lung cancer patients. METHODS One hundred lung cancer patients who were treated in our hospital from May 2021 to May 2023 were selected as the study subjects and divided into chemotherapy group (n = 50) and ICIs group (n = 50), in which the chemotherapy group was given the combined treatment of vincristine and cisplatin (NP), while the ICIs group was given ICIs for treatment. The therapeutic effect and adverse reactions (hypertriglyceridemia, anemia, hypertension and hypoproteinemia) of the two groups were compared, and fasting venous blood was collected. The levels of carcinoembryonic antigen (CEA) and cancer antigen 199 (CA199) were compared between the two groups before and after treatment. According to the therapeutic effect, 100 patients with lung cancer were divided into complete remission (CR) + partial remission (PR) group (n = 52) and stable (SD) + progressive (PD) group (n = 48). The clinical data and pathologic data of the two groups were compared. RESULTS The rates of objective effective rate (ORR) in chemotherapy group and ICIs group were 36.00% and 68.00% respectively, and the level of ORR in ICIs group was significantly higher than that in chemotherapy group, with statistical significance (P < 0.05). There was no significant difference in serum CEA and CA199 levels between the two groups before operation (P > 0.05). Three months after operation, the serum CEA and CA199 levels in ICIs group were significantly lower than those in chemotherapy group, and the difference was statistically significant (P < 0.05). The adverse reactions of hypertriglyceridemia, anemia, hypertension and hypoproteinemia in chemotherapy group and ICIs group during treatment were all grade 1-2, and the incidence of adverse reactions was similar between the two groups (P > 0.05). There was no significant difference in sex, age, anatomic position, pathologic type, smoking history and differentiation between the two groups (P > 0.05). In SD + PD group, the preoperative maximum tumor diameter > 4 cm, tumor node metastasis (TNM) stage IV, lactate dehydrogenase (LDH) ≥ 183 U/L, and tumor volume ≥ 120m3 were significantly higher than those in CR + PR group, and the prognostic nutritional index (PNI) ≥ 41.8 and the proportion of ICIs were significantly lower than those in CR + PR group, with statistical significance (P < 0.05). Multifactorial logistic regression analysis showed that preoperative maximum tumor diameter > 4 cm and LDH ≥ 183 U/L were risk factors for poor lung cancer outcome, and PNI ≥ 41.8 and ICIs treatment were protective factors for poor lung cancer outcome (P < 0.05). CONCLUSION ICIs is effective in the treatment of lung cancer, which can obviously reduce the tumor load and has high safety. In addition, the maximum tumor diameter and LDH are risk factors that affect the poor curative effect of lung cancer. High PNI level and ICIs treatment are helpful to improve the effect of lung cancer, and early monitoring is helpful to guide the treatment plan and evaluate the treatment effect.
Collapse
Affiliation(s)
- Jun Luo
- Department of Laboratory Medicine, Chengdu Second People's Hospital, Chengdu, 610017, China
| | - Li Li
- Department of Respiratory and Critical Care Medicine, Dujiangyan People's Hospital, No. 622, Baolian Road, Dujiangyan, Chengdu, 610017, Sichuan, China.
| | - HongGui Wang
- Department of Pharmacy, Dujiangyan People's Hospital, Dujiangyan, 611830, China
| | - Xian Zhang
- Department of Laboratory Medicine, Chengdu Second People's Hospital, Chengdu, 610017, China
| | - FangTing He
- Department of Laboratory Medicine, Chengdu Second People's Hospital, Chengdu, 610017, China
| | - Meng Shi
- Department of Laboratory Medicine, Chengdu Second People's Hospital, Chengdu, 610017, China
| | - Xin Zhang
- Department of Laboratory Medicine, Chengdu Second People's Hospital, Chengdu, 610017, China
| | - Rui Tang
- Department of Laboratory Medicine, Chengdu Second People's Hospital, Chengdu, 610017, China
| | - Yong Bao
- Department of Laboratory Medicine, Chengdu Second People's Hospital, Chengdu, 610017, China
| |
Collapse
|
2
|
Wei X, Xiang X, Wang H, Wang Z, Xing S, Peng W, Ye L, Qu Y, Chen L, Yang B, Zhang S, Xue Q, Ai J, Jiang K, Zhou Q. Tumor cell-intrinsic circular RNA circFNDC3B attenuates CD8 + T cells infiltration in non-small cell lung cancer. Commun Biol 2025; 8:711. [PMID: 40341878 PMCID: PMC12062398 DOI: 10.1038/s42003-025-08108-6] [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: 06/30/2024] [Accepted: 04/18/2025] [Indexed: 05/11/2025] Open
Abstract
Tumor-infiltrating CD8+ T cells are critical for anti-tumor immunity and positively associated with patient survival. However, the mechanisms governing CD8+ T cell infiltration remain incompletely elucidated, particularly those involving circular RNAs (circRNAs). In this study, we characterized circRNA expression profiles in four paired normal and tumor tissues of non-small-cell lung cancer (NSCLC) and identified that circFNDC3B, a circular transcript derived from exons 2 and 3 of the fibronectin type III domain containing 3B (FNDC3B) gene, as significantly upregulated in NSCLC tissues. Mechanistic investigations revealed that circFNDC3B directly binds to transcription factor II-I (TFII-I), forming an RNA-protein complex that competitively disrupts the interaction between TFII-I and STAT1. This sequestration abrogates the transcriptional activation of CXCL10 and CXCL11, two critical chemokines governing CD8+ T cell chemoattraction. Consequently, reduced CXCL10/11 expression significantly impairs CD8+ T cell infiltration into the tumor microenvironment. Consistently, the murine ortholog circFndc3b expression exhibits an inverse correlation with CD8+ T cell infiltration in tumors. Our study uncovers a crucial circRNA-mediated regulatory axis wherein circFNDC3B impedes anti-tumor immunity by suppressing chemokine-dependent CD8+ T cell recruitment, positioning circFNDC3B as a potential therapeutic target to enhance CD8+ T cell-mediated anti-tumor responses in NSCLC.
Collapse
MESH Headings
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- Humans
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Animals
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice
- Gene Expression Regulation, Neoplastic
- Tumor Microenvironment/immunology
- Cell Line, Tumor
- Female
Collapse
Affiliation(s)
- Xiaoshan Wei
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Xiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haolei Wang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihao Wang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shijie Xing
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenbei Peng
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linlin Ye
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Qu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long Chen
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bohan Yang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Siyu Zhang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianqian Xue
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaqi Ai
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Jiang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qiong Zhou
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Qureshi Z, Zaheer Z, Asghar Z, Bakhtiar M, Fatima E, Altaf F. Cardiovascular Risk Profile of Nivolumab Anti-cancer Therapy: A Review of Current Literature. Am J Clin Oncol 2025; 48:235-241. [PMID: 40008416 DOI: 10.1097/coc.0000000000001166] [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: 02/27/2025]
Abstract
OBJECTIVES Immune checkpoint inhibitors (ICI) upregulate host antitumor immunity, proving efficacy across diverse tumor types. Currently approved ICI treatment primarily targets the programmed cell death receptor 1 (PD-1) and its ligand PD-L1, and cytotoxic T lymphocyte-antigen 4 (CTLA-4). Nivolumab is a monoclonal antibody that targets the human PD-1 receptor and is an entirely human immunoglobulin G4 (IgG4), approved by the FDA for various cancers like advanced melanoma, metastatic renal cell carcinoma, Hodgkin lymphoma, and advanced lung carcinoma. This review will summarise and discuss the recent literature on cardiotoxicity associated with nivolumab therapy. METHODS We searched online databases like PubMed, Scopus, Google Scholar, and Embase for articles related to Nivolumab. RESULTS Cardiotoxicity with ICI use is most commonly represented as myocarditis. Patients present with complaints of shortness of breath, palpitations, edema, and fatigue. Takotsubo cardiomyopathy, or broken heart syndrome, is characterized by systolic dysfunction of the left ventricle, mimicking a myocardial infarction but without associated coronary ischemia and with minimal elevation of cardiac enzymes. In the CHECKMATE-037 trial, ventricular arrhythmias occurred in <10% of those who received nivolumab. In a retrospective analysis of patients treated with ICI (predominantly nivolumab monotherapy) for lung cancer, 11% of the patients developed major adverse cardiac events, including myocarditis, non-ST-segment elevated myocardial infarction, supraventricular tachycardia, and pericardial disorders. CONCLUSION Close collaboration between cardiology and oncology specialists is crucial for early detection and effective management of cardiac complications, enhancing the safety of nivolumab anticancer therapy.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | | | - Zoha Asghar
- Department of Medicine, Ziauddin University, Karachi
| | | | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| |
Collapse
|
4
|
Lee J, Kang Y, Lee H, Saravanakumar G, Park SA, Ahn S, Kim WJ. Amplifying glioblastoma immunotherapy: T cell shielding through Nitric oxide/reactive oxygen species scavenging nanoparticles Potentiates anti-PD-1. Biomaterials 2025; 315:122904. [PMID: 39490061 DOI: 10.1016/j.biomaterials.2024.122904] [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: 09/02/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 11/05/2024]
Abstract
Despite the success of immune checkpoint blockade (ICB) therapy in various cancers, its efficacy faces challenges in glioblastoma (GBM) due to the immunosuppressive cold-tumor microenvironment. The scarcity of tumor-infiltrating T cells and the suppression of T cell activity significantly limit therapeutic outcomes in GBM. Nitric oxide (NO) and reactive oxygen species (ROS) from tumor-associated myeloid cells (TAMCs) are key contributors to T cell suppression, reducing ICB therapy effectiveness. In this study, we developed NO-ROS scavenging micelles that effectively scavenge both NO and ROS, protecting T cells from their exhausting effects. This leads to a significant increase in T cell infiltration and activation. Moreover, when combined with αPD-1, the survival rate increases to 40 % up to 120 days, enhancing therapeutic efficacy compared to αPD-1 alone. This approach not only protects T cells from the inhibitory effects of NO and ROS but also has the potential to reshape the tumor microenvironment, overcoming T cell suppression in cold tumors.
Collapse
Affiliation(s)
- Jihye Lee
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Yeoul Kang
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Hyori Lee
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | | | - Soon A Park
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Stephen Ahn
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Won Jong Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea; OmniaMed Co., Ltd, Pohang, 37666, Republic of Korea.
| |
Collapse
|
5
|
Apter L, Sharman Moser S, Gazit S, Chodick G, Hoshen M, Greenberg D, Siegelmann-Danieli N. Healthcare resource utilization and associated cost in patients with metastatic non-small cell lung cancer treated in the immunotherapy era. Oncologist 2025; 30:oyae240. [PMID: 39340826 PMCID: PMC11954502 DOI: 10.1093/oncolo/oyae240] [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: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Treatment approach for metastatic non-small cell lung cancer (mNSCLC) has revolutionized in the recent decade with the introduction of immunotherapy and targeted medications in first-line (1L) therapy. We present real-world data on clinical outcomes and direct healthcare resource utilization (HCRU) and cost in a 2.7-million-member Israeli health provider. PATIENTS AND METHODS Newly diagnosed mNSCLC patients between January 2017 and December 2020 were categorized by 1L treatment: platinum-based chemotherapy, targeted therapy, or immunotherapy. HCRU and costs were calculated based on the Ministry of Health Prices and were assessed at a minimum of 6 months' follow-up (cutoff: 30 June 2021). RESULTS A total of 886 patients were included in the study: 40.6% female, median age 68 years (IQR 61-74), 24.3% never smokers, 80.6% with adenocarcinoma, and 54% with a 0-1 performance status. The median follow-up was 27.12 months (95% CI, 24.7-29.6) and the median duration of first-line (1L) treatment was 2.3 months for platinum-based chemotherapy (n = 177), 12.3 months for targeted therapy (n = 255), and 4.8 months for immunotherapy (n = 463). The median overall survival was 9.09, 27.68, and 12.46 months, respectively. Total 1L costs were driven by radiotherapy for platinum-based chemotherapy and medication for targeted therapy or immunotherapy. Total costs for deceased patients over the entire follow-up were €121 155, €129 458, and €110 716, respectively. CONCLUSION The treatment of mNSCLC carries a high economic burden, primarily driven by first-line therapy, especially with targeted and immune therapies. Further studies are needed to evaluate the impact of innovative treatments on the disease management costs of mNSCLC.
Collapse
Affiliation(s)
- Lior Apter
- Health Division, Maccabi Healthcare Services, Tel Aviv 6812509, Israel
- Department of Health Policy and Management, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be’er-Sheva 8410501, Israel
| | - Sarah Sharman Moser
- Maccabi Institute for Research and Innovation (Maccabitech), Maccabi Healthcare Services, Tel Aviv 6812509, Israel
| | - Sivan Gazit
- Maccabi Institute for Research and Innovation (Maccabitech), Maccabi Healthcare Services, Tel Aviv 6812509, Israel
| | - Gabriel Chodick
- Maccabi Institute for Research and Innovation (Maccabitech), Maccabi Healthcare Services, Tel Aviv 6812509, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Moshe Hoshen
- Maccabi Institute for Research and Innovation (Maccabitech), Maccabi Healthcare Services, Tel Aviv 6812509, Israel
| | - Dan Greenberg
- Department of Health Policy and Management, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be’er-Sheva 8410501, Israel
| | - Nava Siegelmann-Danieli
- Health Division, Maccabi Healthcare Services, Tel Aviv 6812509, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
6
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
7
|
Gao R, Liang W, Chen J, Yang M, Yu X, Wang X. Comparisons of adverse events associated with immune checkpoint inhibitors in the treatment of non-small cell lung cancer: a real-world disproportionality analysis based on the FDA adverse event reporting system. BMC Cancer 2025; 25:216. [PMID: 39920614 PMCID: PMC11806835 DOI: 10.1186/s12885-025-13614-1] [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: 06/27/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI) therapy is increasingly used to treat non-small cell lung cancer (NSCLC). However, little attention has been given to the comparative analysis of adverse events (AEs) associated with different ICIs. METHODS Disproportionality analysis and Bayesian confidence propagation neural network (BCPNN) were utilized to identify pharmacovigilance signals from the FDA Adverse Event Reporting System (FAERS). We compared the sex distribution of patients, risk of suffering more severe adverse reactions, and risk of hospitalization associated with different ICIs, using pairwise matrices that displayed odds ratio (OR) and their 95% confidence interval (CI). And we also compared the outcomes of reactions by using ordinal logistic regression. RESULTS We analyzed 13,580 reports of AEs associated with five ICIs, namely, durvalumab, pembrolizumab, ipilimumab, atezolizumab, and nivolumab from January 2013 to October 2022. Significant differences were observed in sex distribution of patients, risk of suffering more severe adverse reactions, risk of hospitalization, and the outcomes of reactions. In terms of respiratory AEs, pembrolizumab exhibited a higher risk compared to durvalumab (OR = 2.48, 95% CI: 1.72-3.59), atezolizumab (OR = 1.84, 95% CI: 1.07-3.16), and nivolumab (OR = 4.21, 95% CI: 1.72-10.28), while ipilimumab exhibited a higher risk compared to durvalumab (OR = 2.76, 95% CI: 1.14-6.65) and nivolumab (OR = 4.67, 95% CI: 1.14-15.51). In terms of endocrine and metabolic AEs, durvalumab (OR = 7.80, 95% CI: 1.33-45.90) and nivolumab (OR = 5.20, 95% CI: 1.17-23.03) exhibited a higher risk compared to ipilimumab. CONCLUSION Each ICI has distinctive features of pharmacovigilance signals. It is essential to acknowledge the AEs associated with the relevant system when clinicians administer ICIs.
Collapse
Affiliation(s)
- Ruichen Gao
- Department of Pulmonary and Critical Care Medicine, Affiliated Changzhou Second Hospital of Nanjing Medical University, 468 Yanling Middle Road, Changzhou, Jiangsu, 213000, China
| | - Wenjun Liang
- Department of Pulmonary and Critical Care Medicine, Affiliated Changzhou Second Hospital of Nanjing Medical University, 468 Yanling Middle Road, Changzhou, Jiangsu, 213000, China
| | - Jintao Chen
- Department of Pulmonary and Critical Care Medicine, Affiliated Changzhou Second Hospital of Nanjing Medical University, 468 Yanling Middle Road, Changzhou, Jiangsu, 213000, China
| | - Mingxia Yang
- Department of Pulmonary and Critical Care Medicine, Affiliated Changzhou Second Hospital of Nanjing Medical University, 468 Yanling Middle Road, Changzhou, Jiangsu, 213000, China
| | - Xiaowei Yu
- Department of Pulmonary and Critical Care Medicine, Affiliated Changzhou Second Hospital of Nanjing Medical University, 468 Yanling Middle Road, Changzhou, Jiangsu, 213000, China
| | - Xiaohua Wang
- Department of Pulmonary and Critical Care Medicine, Affiliated Changzhou Second Hospital of Nanjing Medical University, 468 Yanling Middle Road, Changzhou, Jiangsu, 213000, China.
| |
Collapse
|
8
|
Yang D, Karanth SD, Yoon HS, Yang JJ, Lou X, Bian J, Zhang D, Guo Y, Yaghjyan L, Akinyemiju T, Rodriguez E, Mehta HJ, Braithwaite D. Disparities in Utilization of Immune Checkpoint Inhibitor Therapy Among Older Patients With Advanced Non-Small Cell Lung Cancer: A SEER-Medicare Analysis. JCO ONCOLOGY ADVANCES 2024; 1:e2400008. [PMID: 39758136 PMCID: PMC11698018 DOI: 10.1200/oa.24.00008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/30/2024] [Accepted: 10/28/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE In the United States, there are disparities in access to care for patients with non-small cell lung cancer (NSCLC) on the basis of socioeconomic and racial/ethnic factors. This study investigates the association between race/ethnicity and the utilization of immune checkpoint inhibitor (ICI) therapy among older patients with advanced NSCLC (aNSCLC). METHODS This retrospective study used data from the SEER-Medicare-linked database. The cohort included patients (age 66 years or older) diagnosed with aNSCLC (stage III/IV) between March 2015 and December 2017, and they were followed through December 2019. Race/ethnicity was categorized as non-Hispanic (NH)-White, NH-Black, Hispanic, and Other. ICI therapy utilization was determined by identifying any usage of ICI agents (nivolumab, pembrolizumab, atezolizumab, durvalumab, ipilimumab, and cemiplimab-rwlc) from the Medicare database. Multivariable logistic regression models assessed the association between race/ethnicity and ICI therapy utilization (yes, no). Effect measure modification analyses were conducted by sex, socioeconomic status, and comorbidity. RESULTS The final sample included 26,836 patients; 76.2% were NH-White, 10.1% NH-Black, 5.7% Hispanic, and 8.0% Other. The overall ICI therapy utilization proportion was 17.8%, varying across ethnicities: NH-Black 14.1%, Hispanic 16.3%, NH-White 18.4%, and Other 18.5%. In comparison with NH-White patients, NH-Black patients were 15% less likely to receive ICI therapy (adjusted odds ratio, 0.85 [95% CI, 0.75 to 0.96]). Furthermore, the association between race/ethnicity and utilization of ICI therapy was modified by comorbidity status, sex, and socioeconomic status. CONCLUSION NH-Black patients with aNSCLC were less likely to receive ICI therapy than their NH-White counterparts. Our findings indicate the racial/ethnic disparities in ICI therapy utilization and call for further interventions to optimize access to care.
Collapse
Affiliation(s)
- Danting Yang
- Department of Epidemiology, University of Florida College of Public Health and Health Professions, Gainesville, FL
| | - Shama D. Karanth
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Hyung-Suk Yoon
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Jae Jeong Yang
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Xiwei Lou
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL
| | - Jiang Bian
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL
| | | | - Yi Guo
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL
| | - Lusine Yaghjyan
- Department of Epidemiology, University of Florida College of Public Health and Health Professions, Gainesville, FL
| | - Tomi Akinyemiju
- Department of Population Health Sciences, School of Medicine, Duke University, Durham, NC
- Duke Cancer Institute, School of Medicine, Duke University, Durham, NC
| | | | - Hiren J. Mehta
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL
| | - Dejana Braithwaite
- Department of Epidemiology, University of Florida College of Public Health and Health Professions, Gainesville, FL
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| |
Collapse
|
9
|
Ulas A, Temel B, Kos FT. Comparison of Prognostic Values of Seven Immune Indexes in Advanced Non-Small-Cell Lung Cancer Treated with Nivolumab: How Effective Can They Be Regarding Our Treatment Decisions? MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1792. [PMID: 39596977 PMCID: PMC11596302 DOI: 10.3390/medicina60111792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/27/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives: In this study, we evaluated the impact of seven immune indexes on treatment response and survival outcomes in advanced non-small-cell lung cancer (NSCLC) patients receiving second-line and subsequent nivolumab treatment under real-life conditions. Materials and Methods: The pan-immune inflammation value (PIV), systemic immune inflammation value (SII), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR), derived neutrophil-to-lymphocyte ratio (d-NLR), and prognostic nutritional index (PNI) were calculated. All immune indexes were classified as low and high based on cut-off values. Kaplan-Meier and Cox hazard models were used for survival analysis. Results: The median follow-up was 22.0 months (6.0-96.0). The median overall survival (OS) was 30.0 months and the median progression-free survival (PFS) was 7.0 months. In the univariate analysis, comorbidity (p = 0.03) and nivolumab use for more than eight cycles (p < 0.0001) were associated with an increase in PFS, while smoking history (p < 0.005) and d-NLR (p < 0.05) were more effective regarding OS. Patients who received more than eight cycles of nivolumab had longer median PFS (4 vs. 19 months, p < 0.001) and OS (23 vs. 43 months, p < 0.001). We found longer median OS in the PLR (45.7 vs. 75.4 months; p = 0.05), PIV (53.0 vs. 66.4 months; p = 0.19), SII (50.0 vs. 71.9 vs. months, p = 0.19), and NLR (49.9 vs. 74.55 months, p = 0.10) indexes in nivolumab long-term users (high vs. low groups, respectively). In short-term users of nivolumab, only d-NLR median OS (high vs. low, 19 vs. 75.2 months, p = 0.07) was different. Complete and partial response rates to nivolumab treatment were higher in the PNI-high group (p = 0.04). Conclusions: In these real-life data, we determined that the PLR, PIV, SII, and NLR indexes were effective in the prognosis of patients who received PD1 inhibitor nivolumab for a long time, and the d-NLR index was effective in those who developed progression in a short time. We found that the PNI was effective in patients who responded well to ICI treatment.
Collapse
Affiliation(s)
- Arife Ulas
- Department of Medical Oncology, University of Health Sciences, Bursa City Education and Research Hospital, 16059 Bursa, Turkey
| | - Beyza Temel
- Department of Internal Medicine, University of Health Sciences, Bursa City Education and Research Hospital, 16059 Bursa, Turkey;
| | - Fahriye Tugba Kos
- Department of Medical Oncology, University of Health Sciences, Ankara City Education and Research Hospital, 06290 Ankara, Turkey;
| |
Collapse
|
10
|
Apter L, Sharman Moser S, Arunachalam A, Gazit S, Hoshen M, Chodick G, Siegelmann-Danieli N. Real-world treatment patterns, biomarker testing, and clinical outcomes of metastatic non-small cell lung cancer patients in the immunotherapy era. Front Oncol 2024; 14:1442909. [PMID: 39512773 PMCID: PMC11543355 DOI: 10.3389/fonc.2024.1442909] [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: 06/03/2024] [Accepted: 10/01/2024] [Indexed: 11/15/2024] Open
Abstract
Background Treatment for first-line (1L) metastatic non-small cell cancer (mNSCLC) changed with the introduction of immunotherapy. We describe treatment utilization and clinical outcomes in a real-world mNSCLC cohort in a 2.7-million-member state-mandated health provider. Methods Newly diagnosed mNSCLC patients initiating systemic anti-cancer treatment (January 2017-December 2020) were identified from the National Cancer Registry. Real-world time on treatment (rwToT) was defined as the length of time between the first and last administration date of treatment. Real-world overall survival (rwOS) was estimated using Kaplan-Meier analysis. Outcomes were assessed at a minimum of 6 months' follow-up (cutoff: 30 June 2021). Results Among 843 patients, 85% had adenocarcinoma (NSQ) and 15% had squamous cell carcinoma (SQ) histology: of these, 43% and 26% were women, median age was 67 and 69 years, and 55% and 48% had 0-1 ECOG performance status, respectively (missing: 27% and 30%, respectively). Median follow-up for the entire cohort was 27.1 months (95% CI: 24.7-29.6). NSQ patients with no known EGFR/ALK/ROS1 aberrations received PD-1 inhibitor monotherapy (PDM) (N = 147) or combination (PDC) (N = 194) or platinum-based chemotherapy (PBC, N = 133). Median rwToT was 4.5 (95% CI: 3.5-7.6), 5.2 (95% CI: 4.6-7.6), and 2.3 (95% CI: 2.1-3.0) months, respectively; for the subgroup of patients with ECOG PS 0-1, rwToT was 9.4 (95% CI: 5.0-20.8), 7.1 (95% CI: 5.0-10.1), and 2.9 (95% CI: 2.2-4.1) months, respectively. Median rwOS from 1L was 12.5 (95% CI: 9.9-17.9), 14.8 (95% CI: 10.5-19.4), and 9.1 (95% CI: 7.1-11.5) months; for the subgroup of patients with ECOG PS 0-1, median rwOS was 25.1 [95% CI: 14.9-not reached (NR)], 17.6 (95% CI: 14.3-NR), and 11.3 (95% CI: 9.2-21.3) months, respectively. For ECOG PS 0-1 and PD-L1 ≥50% patients, median rwOS was 25.1 months (95% CI: 13.9-NR) and NR for PDM and PDC, respectively. For ECOG PS 0-1 and PD-L1 <50% patients, median rwOS was 14.3 (95% CI: 10.1-NR) and 11.2 (95% CI: 9.1-21.3) months for PDC and PBC, respectively. Conclusion Our real-world data support the benefit of single-agent PD-1 inhibitor monotherapy for patients with PD-L1 high expression or PD-1 inhibitor combination for all patients diagnosed with mNSCLC with no known EGFR/ALK/ROS1 aberrations, initiating 1L treatment.
Collapse
Affiliation(s)
- Lior Apter
- Health Division, Maccabi Healthcare Services, Tel Aviv, Israel
- Department of Health Policy and Management, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be’er-Sheva, Israel
| | - Sarah Sharman Moser
- KSM Maccabi Institute for Research and Innovation, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Ashwini Arunachalam
- Outcomes Research, Value & Implementation, Merck & Co., Inc., Rahway, NJ, United States
| | - Sivan Gazit
- KSM Maccabi Institute for Research and Innovation, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Moshe Hoshen
- KSM Maccabi Institute for Research and Innovation, Maccabi Healthcare Services, Tel Aviv, Israel
| | | | - Nava Siegelmann-Danieli
- Health Division, Maccabi Healthcare Services, Tel Aviv, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
11
|
Cui S, Yang Y, Lou S, Huang R, Wang J, Chen Z, Xie J. Establish a novel immune-related gene prognostic risk index (IRGPRI) associated with CD8+ cytotoxic T lymphocytes in non-small-cell lung cancer (NSCLC). Heliyon 2024; 10:e38324. [PMID: 39397989 PMCID: PMC11466668 DOI: 10.1016/j.heliyon.2024.e38324] [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: 04/22/2024] [Revised: 09/22/2024] [Accepted: 09/22/2024] [Indexed: 10/15/2024] Open
Abstract
Background The aim of this study is to create an index called IRGPRI (immune-related gene prognostic risk index) that can be utilized for predicting the prognosis and assessing the efficacy of immune checkpoint inhibitors (ICIs) therapy in patients with non-small-cell lung cancer (NSCLC). Methods Distinguishing gene expression patterns (DEGs) were detected in CD8+ cytotoxic T lymphocytes (CTLs) compared to other cellular types such as CD4 T cells, B cells, plasma cells, and CD8 Tex using the advanced technology of Single-cell RNA Sequencing (scRNA-seq). The construction of IRGPRI was accomplished by employing LASSO Cox regression analysis. We conducted a comparative analysis on clinical characteristics and molecular features, such as pathway enrichment and gene mutation, among the distinct subgroups of IRGPRI. Furthermore, we explored the correlation between immunological characteristics and IRGPRI subgroups to comprehensively assess the effectiveness of ICIs in NSCLC patients. Results A total of 109 genes were identified by intersecting immune-related genes with DEGs obtained from single-cell RNA sequencing data (GSE131907), specifically comparing CTLs to other cell types. From these, we selected 7 prognosis-related genes, namely TRBC1, HLA-DMA, CTSH, RAC1, CTSL, ANXA2, and CEBPB. These genes were used to construct the IRGPRI. The prognosis of patients diagnosed with NSCLC was found to be significantly better in the low-risk group compared to the high-risk group, as demonstrated by Kaplan-Meier (K-M) survival analysis. This observation was further confirmed through the utilization of data from the GEO cohort. The low-risk group demonstrated an increase in pathways linked with immune response, whereas the high-risk group exhibited a higher prevalence of pathways related to cancer. Furthermore, it was noted in the TCGA cohort that there existed a significant rise in the mutation frequency of every gene within the high-risk group as opposed to the low-risk group. Missense variation emerged as the most prevalent form of mutation. According to the analysis of immune cell infiltration and function, the comprehensive findings suggest that the group with a low risk is characterized by an increased presence of plasma cells, CTLs, T cells follicular helper, Tregs, and Dendritic cell resting. Additionally, they exhibit a higher score in terms of immune function for B cells, CD8+ T cells, checkpoint activity, T cell inhibition and stimulation. Moreover, this low-risk group demonstrates greater efficacy when treated with ICIs therapy compared to the high-risk group. Conclusions Our research effectively developed and verified a unique IRGPRI, showcasing its association with immune-related characteristics. As a result, the potential of IRGPRI as a valuable biomarker for predicting prognosis and evaluating the effectiveness of ICIs treatment in cancer is evident.
Collapse
Affiliation(s)
- Shenjing Cui
- Department of Clinical Laboratory, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yikun Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Shuang Lou
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Rong Huang
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jing Wang
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhongbiao Chen
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jingjing Xie
- Department of Medical Administration, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
12
|
Uprety D, Seaton R, Hadid T, Mamdani H, Sukari A, Ruterbusch JJ, Schwartz AG. Racial and socioeconomic disparities in survival among patients with metastatic non-small cell lung cancer. J Natl Cancer Inst 2024; 116:1697-1704. [PMID: 38830035 PMCID: PMC11461161 DOI: 10.1093/jnci/djae118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/25/2024] [Accepted: 05/26/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors have profoundly impacted survival among patients with metastatic non-small cell lung cancer. However, population-based studies evaluating this impact on survival by race and socioeconomic factors are lacking. METHODS We used the Surveillance, Epidemiology, and End Results Program-Medicare database to identify patients with metastatic non-small cell lung cancer diagnosed between 2015 and 2019. The primary study outcomes were the receipt of an immune checkpoint inhibitor and overall survival. χ2 tests and logistic regression were used to identify demographic factors associated with receipt of immune checkpoint inhibitors. The Kaplan-Meier method was used to calculate 2-year overall survival rates, and log-rank tests were used to compare survival by race and ethnicity. RESULTS Of 17 134 patients, approximately 39% received an immune checkpoint inhibitor. Those diagnosed with cancer recently (in 2019); who are relatively younger (aged younger than 85 years); non-Hispanic White, non-Hispanic Asian, or Hispanic; living in high socioeconomic status or metropolitan areas; not Medicaid eligible; and with adenocarcinoma histology were more likely to receive immune checkpoint inhibitors. The 2-year overall survival rate from diagnosis was 21% for the overall population. The 2-year overall survival rate from immune checkpoint inhibitor initiation was 30%, among those who received at least 1 cycle and 11% among those who did not receive immune checkpoint inhibitors. The 2-year overall survival rates were higher among non-Hispanic White (22%) and non-Hispanic Asian (23%) patients compared with non-Hispanic Black (15%) and Hispanic (17%) patients. There was no statistically significant racial differences in survival for those who received immune checkpoint inhibitors. CONCLUSION Immune checkpoint inhibitor utilization rates and the resulting outcomes were inferior for certain vulnerable groups, mandating the need for strategies to improve access to care.
Collapse
Affiliation(s)
- Dipesh Uprety
- Department of Medical Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Randell Seaton
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Tarik Hadid
- Department of Medical Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Hirva Mamdani
- Department of Medical Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Ammar Sukari
- Department of Medical Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Julie J Ruterbusch
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Ann G Schwartz
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| |
Collapse
|
13
|
Wei Y, Zhang R, Yin R, Wang S, Han J, Chen R, Fu Z. Immunotherapy Improves the Survival of Stage 4 Non-Small Cell Lung Cancer Patients at the US Population Level: The Real-World Evidence. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e70000. [PMID: 39275901 PMCID: PMC11399776 DOI: 10.1111/crj.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 09/16/2024]
Abstract
INTRODUCTION Immunotherapy has revolutionized the management of lung cancer and improved lung cancer survival in trials, but its real-world impact at the population level remains unclear. METHODS Using data obtained from eight Surveillance, Epidemiology, and End Results (SEER) registries from 2004 through 2019, we addressed the long-term trends in the incidence, incidence-based mortality (IBM), and survival of lung cancer patients in the United States. RESULTS The incidence and IBM of both non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) all significantly decreased steadily from 2004 to 2019. The 1-year survival (1-YS) of both NSCLC and SCLC improved over time, with the best improvement observed for Stage 4 NSCLC. Two significant turning points of Stage 4 NSCLC 1-YS were observed over the years: 0.63% (95% confidence interval [CI]: 0.33%-0.93%) from 2004 to 2010, 0.81% (95% CI: 0.41%-1.21%) from 2010 to 2014 and a striking 2.09% (95% CI: 1.70%-2.47%) from 2014 to 2019. The same two turning points in 1-YS were pronounced for Stage 4 NSCLC in women, which were coincident with the introduction of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) and immunotherapy. However, for Stage 4 NSCLC in men, only one significant turning point in the 1-YS starting in 2014 was found, which might only correspond to immunotherapy. Significant period effects in reduced IBM were also observed for both Stage 4 AD and Stage 4 SQCC during the period. CONCLUSION This SEER analysis found that immunotherapy improved the survival of Stage 4 NSCLC patients at the population level in the United States. This real-world evidence confirms that immunotherapy has truly revolutionized the management of lung cancer.
Collapse
Affiliation(s)
- Yuxuan Wei
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Zhang
- College of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Ruikang Yin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shijie Wang
- Radiation Oncology Department, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianglong Han
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruyan Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhenming Fu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
14
|
Graham KL, Carty D, Poulter SP, Blackman C, Dunstan OG, Milton TL, Ferguson C, Smith K, Van Dijk E, Jongebloed D, Loeliger J, Baguley BJ. The nutrition-related adverse events associated with immune checkpoint inhibitor treatment for patients with non-small cell lung cancer: A systematic review. Nutr Diet 2024; 81:356-370. [PMID: 38804022 DOI: 10.1111/1747-0080.12879] [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: 10/12/2023] [Revised: 03/24/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024]
Abstract
AIMS Immune checkpoint inhibitor therapy used for lung cancer has significantly changed response and survival rates, however, the impact on patients' nutritional status remains largely unexplored. This review aims to identify common adverse events that increase nutrition risk induced in non-small cell lung cancer patients treated with immune checkpoint inhibitor therapy and assess impact on nutritional status. METHODS PubMed, Medline and CINAHL were systematically searched in September 2023 for randomised controlled trials comparing immune checkpoint inhibitor treatment of non-small cell lung cancer to a control group. Treatment-related adverse events that increased nutrition impact symptoms identified in the patient-generated subjective global assessment and clinical guidelines were extracted and qualitatively analysed. Risk of bias was assessed using Cochrane Risk of Bias tool 2. RESULTS Eleven eligible randomised controlled trial studies were identified and analysed. The data demonstrated immune checkpoint inhibitor treatment was associated with a lower percentage of reported nutrition impact symptoms, for example, decreased appetite, nausea, vomiting, compared to chemotherapy treatment. Conversely, immune checkpoint inhibitor treated patients recorded a greater percentage of immune-related adverse events that alter metabolism or nutrient absorption. CONCLUSION Non-small cell lung cancer patients treated with immune checkpoint inhibitors still experience nutrition impact symptoms but less frequently than patients treated with chemotherapy. This combined with unique nutrition-related consequences from colitis and thyroid disorders induced by immune checkpoint inhibitor therapy indicates patients should be screened, assessed and interventions implemented to improve nutrition.
Collapse
Affiliation(s)
- Kate L Graham
- Nutrition & Speech Pathology Department, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Danika Carty
- Nutrition & Speech Pathology Department, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Shay P Poulter
- School of Exercise and Nutrition Sciences, Deakin University, Burwood, Victoria, Australia
| | - Chantal Blackman
- School of Exercise and Nutrition Sciences, Deakin University, Burwood, Victoria, Australia
| | - Olivia G Dunstan
- School of Exercise and Nutrition Sciences, Deakin University, Burwood, Victoria, Australia
| | - Taryn L Milton
- School of Exercise and Nutrition Sciences, Deakin University, Burwood, Victoria, Australia
| | - Cassie Ferguson
- School of Exercise and Nutrition Sciences, Deakin University, Burwood, Victoria, Australia
| | - Kaitlyn Smith
- School of Exercise and Nutrition Sciences, Deakin University, Burwood, Victoria, Australia
| | - Emilly Van Dijk
- School of Exercise and Nutrition Sciences, Deakin University, Burwood, Victoria, Australia
| | - Darcy Jongebloed
- Nutrition & Speech Pathology Department, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jenelle Loeliger
- Nutrition & Speech Pathology Department, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Institute for Physical Activity and Nutrition, Deakin University, Geelong, Victoria, Australia
| | - Brenton J Baguley
- School of Exercise and Nutrition Sciences, Deakin University, Burwood, Victoria, Australia
- Institute for Physical Activity and Nutrition, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
15
|
Rina A, Maffeo D, Minnai F, Esposito M, Palmieri M, Serio VB, Rosati D, Mari F, Frullanti E, Colombo F. The Genetic Analysis and Clinical Therapy in Lung Cancer: Current Advances and Future Directions. Cancers (Basel) 2024; 16:2882. [PMID: 39199653 PMCID: PMC11352260 DOI: 10.3390/cancers16162882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Lung cancer, including both non-small cell lung cancer and small cell lung cancer, remains the leading cause of cancer-related mortality worldwide, representing 18% of the total cancer deaths in 2020. Many patients are identified already at an advanced stage with metastatic disease and have a worsening prognosis. Recent advances in the genetic understanding of lung cancer have opened new avenues for personalized treatments and targeted therapies. This review examines the latest discoveries in the genetics of lung cancer, discusses key biomarkers, and analyzes current clinical therapies based on this genetic information. It will conclude with a discussion of future prospects and potential research directions.
Collapse
Affiliation(s)
- Angela Rina
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- UOC Laboratorio di Assistenza e Ricerca Traslazionale, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy;
| | - Debora Maffeo
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Francesca Minnai
- Institute of Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (F.M.); (M.E.)
| | - Martina Esposito
- Institute of Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (F.M.); (M.E.)
| | - Maria Palmieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Viola Bianca Serio
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Diletta Rosati
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Francesca Mari
- UOC Laboratorio di Assistenza e Ricerca Traslazionale, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy;
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Elisa Frullanti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Francesca Colombo
- Institute of Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (F.M.); (M.E.)
| |
Collapse
|
16
|
Rahman M, Akter K, Ahmed KR, Fahim MMH, Aktary N, Park MN, Shin SW, Kim B. Synergistic Strategies for Castration-Resistant Prostate Cancer: Targeting AR-V7, Exploring Natural Compounds, and Optimizing FDA-Approved Therapies. Cancers (Basel) 2024; 16:2777. [PMID: 39199550 PMCID: PMC11352813 DOI: 10.3390/cancers16162777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/29/2024] [Accepted: 08/03/2024] [Indexed: 09/01/2024] Open
Abstract
Castration-resistant prostate cancer (CRPC) remains a significant therapeutic challenge due to its resistance to standard androgen deprivation therapy (ADT). The emergence of androgen receptor splice variant 7 (AR-V7) has been implicated in CRPC progression, contributing to treatment resistance. Current treatments, including first-generation chemotherapy, androgen receptor blockers, radiation therapy, immune therapy, and PARP inhibitors, often come with substantial side effects and limited efficacy. Natural compounds, particularly those derived from herbal medicine, have garnered increasing interest as adjunctive therapeutic agents against CRPC. This review explores the role of AR-V7 in CRPC and highlights the promising benefits of natural compounds as complementary treatments to conventional drugs in reducing CRPC and overcoming therapeutic resistance. We delve into the mechanisms of action underlying the anti-CRPC effects of natural compounds, showcasing their potential to enhance therapeutic outcomes while mitigating the side effects associated with conventional therapies. The exploration of natural compounds offers promising avenues for developing novel treatment strategies that enhance therapeutic outcomes and reduce the adverse effects of conventional CRPC therapies. These compounds provide a safer, more effective approach to managing CRPC, representing a significant advancement in improving patient care.
Collapse
Affiliation(s)
- Muntajin Rahman
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Khadija Akter
- Department of Plasma Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea;
| | - Kazi Rejvee Ahmed
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Md. Maharub Hossain Fahim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Nahida Aktary
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Sang-Won Shin
- Department of Humanities & Social Medicine, School of Korean Medicine, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan-si 50612, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
- Department of Plasma Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea;
| |
Collapse
|
17
|
Liu TC, Shih CJ, Chiou YL. Oral administration of oligo fucoidan improves the survival rate, quality of life, and immunity in patients with lung cancer. Food Nutr Res 2024; 68:10674. [PMID: 38974912 PMCID: PMC11227263 DOI: 10.29219/fnr.v68.10674] [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: 03/25/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 07/09/2024] Open
Abstract
Background Lung cancer, the most commonly diagnosed cancer globally, has the highest incidence and mortality rates in Taiwan. It can be divided into two types. Non-small cell lung cancer (NSCLC) accounts for approximately 85% of lung cancers, which is further divided into adenocarcinoma, squamous cell carcinoma, and large cell lung cancer accounting for approximately 40%, 25%, and 15% of NSCLC cases, respectively. Small cell lung cancer accounts for approximately 15% of lung cancers. Early systemic therapy NSCLC was based on chemotherapy, and immunotherapy is currently under development. Fucoidan, from brown seaweed extracts, shows promise in mitigating radiation-induced lung fibrosis in animal studies, suggesting its potential as an adjuvant for radiation therapy-related lung fibrosis in lung cancer patients. However, the clinical utility of such adjuvant therapy in lung cancer treatment remains uncertain. The purpose of this study was to investigate the effects of oral administration of oligo-fucoidan on the survival rate, quality of life, and immunity of patients with lung cancer. Methods Subjects with Non-small cell lung cancer aged between 20 and 80 were collected from outpatient clinics, divided into control group (n = 7): conventional therapy and fucoidan group (n = 13): received conventional therapy+ oral supplementation of oligo-fucoidan (550 mg × 4 tablets). Data were collected before the study, at weeks 4, 12, and 24 during the study, and to collect 20 ml of peripheral blood, for analysis biochemical data, liver and kidney function, lymphocyte population, inflammation cytokines, and using EORTC QLQ-C30 questionnaire to assess quality of life. Results The survival rates of the subjects in the control and fucoidan groups were 20% and 28.6%, respectively. During the study, patients in the fucoidan group experienced a better quality of life than those in the control group, but this difference lacked statistical significance. Oligo-fucoidan increases the CD19 lymphocyte population. The patients in the fucoidan group also had Lower inflammatory cytokine. Conclusion Oligo-fucoidan holds promise as an adjuvant therapy to enhance the survival rate, quality of life, and immune function in patients with lung cancer.
Collapse
Affiliation(s)
- Tu-Chen Liu
- Department of Chest Medicine, Cheng-Ching General Hospital, Taichung, Republic of China
| | - Chia-Ju Shih
- Department of Nutrition (Master Program), Hungkuang University, Taichung, Taiwan, Republic of China
| | - Ya-Ling Chiou
- Department of Nutrition (Master Program), Hungkuang University, Taichung, Taiwan, Republic of China
| |
Collapse
|
18
|
Karadurmus N, Kaplan MA, Sendur MAN, Urun Y, Demirci U, Karaca SB, Goktas Aydin S, Aykan MB, Bilici A, Sezer A, Yilmaz U, Abali H, Yumuk PF, Degirmencioglu S, Demirkazik A, Paydas S, Mirili C, Turna H, Kargi A, Ozdogan M, Guven DC, Ozguroglu M, Kilickap S. Evaluation of the efficacy and safety of nivolumab in the second- or later-line treatment of patients with locally advanced/metastatic non-small cell lung cancer in Türkiye: a retrospective multicenter non-interventional registry study. Curr Med Res Opin 2024; 40:1171-1178. [PMID: 38809230 DOI: 10.1080/03007995.2024.2359026] [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: 11/21/2023] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 05/30/2024]
Abstract
OBJECTIVE To evaluate the efficacy and safety of nivolumab in the second-line (2L) or later-line (LL) treatment of patients with locally advanced/metastatic non-small cell lung cancer (NSCLC) in real-life setting in Türkiye. METHODS This study was designed as a national, multi-center, retrospective study. The study population was evaluated in two groups for the line of nivolumab therapy: those receiving nivolumab in the 2L (Group 2L) and third-line (3L) or LL (Group 3L/LL). Efficacy was evaluated based on one-year overall survival (OS) and progression-free survival (PFS). Safety was evaluated based on treatment-related adverse events (AEs) and nivolumab discontinuation rate. RESULTS Of 244 patients, 52.9% were in Group 2L and 47.1% were in Group 3L/LL. Demographic and clinical characteristics did not differ between the groups. In Group 2L and Group 3L/LL, one-year OS and PFS rates were 60.8% and 61.4% (p = 0.592) and 31.2% and 21.3% (p = 0.078), respectively. The objective response rate (ORR) was 34.7% in Group 2L and 27.3% in Group 3L/LL (p = 0.262). The percentage of patients reporting at least one AE in Groups 2L and 3L/LL was 34.9% and 43.5%, respectively (p = 0.169). Fatigue was the most common (16.4%) treatment-related AE in each group. The groups were comparable regarding the AE frequency. Nivolumab was discontinued in 61 patients in Group 2L and 53 patients in Group 3L/LL, with the most common reason being disease progression (57.4% and 66.0%, respectively). CONCLUSION Nivolumab is safe and effective in the 2L or 3L/LL treatment of locally advanced/metastatic NSCLC and associated with acceptable AEs in real-life setting.
Collapse
Affiliation(s)
- Nuri Karadurmus
- Medical Oncology Department, University of Health Sciences, Gulhane Training and Research Hospital, Ankara, Türkiye
| | | | | | - Yuksel Urun
- Medical Oncology Division, Department of Internal Diseases, Ankara University Medical Faculty, Ankara, Türkiye
| | - Umut Demirci
- Medical Oncology Unit, Memorial Ankara Hospital, Ankara, Türkiye
| | | | - Sabin Goktas Aydin
- Medical Oncology Unit, Bagcilar Medipol Mega University Hospital, Istanbul, Türkiye
| | - Musa Baris Aykan
- Medical Oncology Department, University of Health Sciences, Gulhane Training and Research Hospital, Ankara, Türkiye
| | - Ahmet Bilici
- Medical Oncology Unit, Bagcilar Medipol Mega University Hospital, Istanbul, Türkiye
| | - Ahmet Sezer
- Medical Oncology Unit, Adana Dr. Turgut Noyan Application and Research Center, Baskent University, Adana, Türkiye
| | - Ulku Yilmaz
- Chest Diseases Unit, Ankara Ataturk Sanatory Education and Research Hospital, Ankara, Türkiye
| | - Huseyin Abali
- Medical Oncology Unit, Acibadem Adana Hospital, Adana, Türkiye
| | - Perran Fulden Yumuk
- Medical Oncology Unit, Marmara University Medical Faculty, Istanbul, Türkiye
| | - Serkan Degirmencioglu
- Department of Internal Diseases, Pamukkale University Medical Faculty, Denizli, Türkiye
| | - Ahmet Demirkazik
- Medical Oncology Division, Department of Internal Diseases, Ankara University Medical Faculty, Ankara, Türkiye
| | - Semra Paydas
- Department of Medical Oncology, Cukurova University Medical Faculty, Adana, Türkiye
| | - Cem Mirili
- Department of Medical Oncology, Cukurova University Medical Faculty, Adana, Türkiye
| | - Hande Turna
- Medical Oncology Division, Department of Internal Medicine, Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Istanbul, Türkiye
| | - Aysegul Kargi
- Medical Oncology Unit, Medstar Antalya Hospital Cancer Center, Antalya, Türkiye
| | - Mustafa Ozdogan
- Medical Oncology Unit, Medstar Antalya Hospital Cancer Center, Antalya, Türkiye
| | - Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Türkiye
| | - Mustafa Ozguroglu
- Medical Oncology Division, Department of Internal Medicine, Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Istanbul, Türkiye
| | | |
Collapse
|
19
|
Tóth LJ, Mokánszki A, Méhes G. The rapidly changing field of predictive biomarkers of non-small cell lung cancer. Pathol Oncol Res 2024; 30:1611733. [PMID: 38953007 PMCID: PMC11215025 DOI: 10.3389/pore.2024.1611733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024]
Abstract
Lung cancer is a leading cause of cancer-related death worldwide in both men and women, however mortality in the US and EU are recently declining in parallel with the gradual cut of smoking prevalence. Consequently, the relative frequency of adenocarcinoma increased while that of squamous and small cell carcinomas declined. During the last two decades a plethora of targeted drug therapies have appeared for the treatment of metastasizing non-small cell lung carcinomas (NSCLC). Personalized oncology aims to precisely match patients to treatments with the highest potential of success. Extensive research is done to introduce biomarkers which can predict the effectiveness of a specific targeted therapeutic approach. The EGFR signaling pathway includes several sufficient targets for the treatment of human cancers including NSCLC. Lung adenocarcinoma may harbor both activating and resistance mutations of the EGFR gene, and further, mutations of KRAS and BRAF oncogenes. Less frequent but targetable genetic alterations include ALK, ROS1, RET gene rearrangements, and various alterations of MET proto-oncogene. In addition, the importance of anti-tumor immunity and of tumor microenvironment has become evident recently. Accumulation of mutations generally trigger tumor specific immune defense, but immune protection may be upregulated as an aggressive feature. The blockade of immune checkpoints results in potential reactivation of tumor cell killing and induces significant tumor regression in various tumor types, such as lung carcinoma. Therapeutic responses to anti PD1-PD-L1 treatment may correlate with the expression of PD-L1 by tumor cells. Due to the wide range of diagnostic and predictive features in lung cancer a plenty of tests are required from a single small biopsy or cytology specimen, which is challenged by major issues of sample quantity and quality. Thus, the efficacy of biomarker testing should be warranted by standardized policy and optimal material usage. In this review we aim to discuss major targeted therapy-related biomarkers in NSCLC and testing possibilities comprehensively.
Collapse
Affiliation(s)
- László József Tóth
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | | |
Collapse
|
20
|
Qin S, Xie B, Wang Q, Yang R, Sun J, Hu C, Liu S, Tao Y, Xiao D. New insights into immune cells in cancer immunotherapy: from epigenetic modification, metabolic modulation to cell communication. MedComm (Beijing) 2024; 5:e551. [PMID: 38783893 PMCID: PMC11112485 DOI: 10.1002/mco2.551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/24/2024] [Accepted: 04/02/2024] [Indexed: 05/25/2024] Open
Abstract
Cancer is one of the leading causes of death worldwide, and more effective ways of attacking cancer are being sought. Cancer immunotherapy is a new and effective therapeutic method after surgery, radiotherapy, chemotherapy, and targeted therapy. Cancer immunotherapy aims to kill tumor cells by stimulating or rebuilding the body's immune system, with specific efficiency and high safety. However, only few tumor patients respond to immunotherapy and due to the complex and variable characters of cancer immune escape, the behavior and regulatory mechanisms of immune cells need to be deeply explored from more dimensions. Epigenetic modifications, metabolic modulation, and cell-to-cell communication are key factors in immune cell adaptation and response to the complex tumor microenvironment. They collectively determine the state and function of immune cells through modulating gene expression, changing in energy and nutrient demands. In addition, immune cells engage in complex communication networks with other immune components, which are mediated by exosomes, cytokines, and chemokines, and are pivotal in shaping the tumor progression and therapeutic response. Understanding the interactions and combined effects of such multidimensions mechanisms in immune cell modulation is important for revealing the mechanisms of immunotherapy failure and developing new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Sha Qin
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of PathologySchool of Basic Medical ScienceXiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Bin Xie
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Qingyi Wang
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of PathologySchool of Basic Medical ScienceXiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Rui Yang
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of PathologySchool of Basic Medical ScienceXiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Jingyue Sun
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of PathologySchool of Basic Medical ScienceXiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Chaotao Hu
- Regenerative Medicine, Medical SchoolUniversity of Chinese Academy of SciencesBeijingChina
| | - Shuang Liu
- Department of OncologyInstitute of Medical SciencesNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha, Hunan, China. UniversityChangshaHunanChina
| | - Yongguang Tao
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of CarcinogenesisCancer Research Institute and School of Basic MedicineCentral South universityChangshaHunanChina
| | - Desheng Xiao
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of PathologySchool of Basic Medical ScienceXiangya School of MedicineCentral South UniversityChangshaHunanChina
| |
Collapse
|
21
|
Lampe H, Tam L, Hansen AR. Bi-specific T-cell engagers (BiTEs) in prostate cancer and strategies to enhance development: hope for a BiTE-r future. Front Pharmacol 2024; 15:1399802. [PMID: 38873417 PMCID: PMC11169794 DOI: 10.3389/fphar.2024.1399802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024] Open
Abstract
Metastatic castrate resistant prostate cancer (mCRPC) continues to have poor survival rates due to limited treatment options. Bi-specific T cell engagers (BiTEs) are a promising class of novel immunotherapies with demonstrated success in haematological malignancies and melanoma. BiTEs developed for tumour associated antigens in prostate cancer have entered clinical testing. These trials have been hampered by high rates of treatment related adverse events, minimal or transient anti-tumour efficacy and generation of high titres of anti-drug antibodies. This paper aims to analyse the challenges faced by the different BiTE therapy constructs and the mCRPC tumour microenvironment that result in therapeutic resistance and identify possible strategies to overcome these issues.
Collapse
Affiliation(s)
| | | | - Aaron R. Hansen
- Department of Medical Oncology, Division of Cancer Care Services, Princess Alexandra Hospital, Metro South Health Service, Queensland Health, Brisbane, QLD, Australia
| |
Collapse
|
22
|
Yu X, Xu J. TWIST1 Drives Cytotoxic CD8+ T-Cell Exhaustion through Transcriptional Activation of CD274 (PD-L1) Expression in Breast Cancer Cells. Cancers (Basel) 2024; 16:1973. [PMID: 38893094 PMCID: PMC11171171 DOI: 10.3390/cancers16111973] [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: 04/12/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
In breast cancer, epithelial-mesenchymal transition (EMT) is positively associated with programmed death ligand 1 (PD-L1) expression and immune escape, and TWIST1 silences ERα expression and induces EMT and cancer metastasis. However, how TWIST1 regulates PD-L1 and immune evasion is unknown. This study analyzed TWIST1 and PD-L1 expression in breast cancers, investigated the mechanism for TWIST1 to regulate PD-L1 transcription, and assessed the effects of TWIST1 and PD-L1 in cancer cells on cytotoxic CD8+ T cells. Interestingly, TWIST1 expression is correlated with high-level PD-L1 expression in ERα-negative breast cancer cells. The overexpression and knockdown of TWIST1 robustly upregulate and downregulate PD-L1 expression, respectively. TWIST1 binds to the PD-L1 promoter and recruits the TIP60 acetyltransferase complex in a BRD8-dependent manner to transcriptionally activate PD-L1 expression, which significantly accelerates the exhaustion and death of the cytotoxic CD8+ T cells. Accordingly, knockdown of TWIST1 or BRD8 or inhibition of PD-L1 significantly enhances the tumor antigen-specific CD8+ T cells to suppress the growth of breast cancer cells. These results demonstrate that TWIST1 directly induces PD-L1 expression in ERα-negative breast cancer cells to promote immune evasion. Targeting TWIST1, BRD8, and/or PD-L1 in ERα-negative breast cancer cells with TWIST1 expression may sensitize CD8+ T-cell-mediated immunotherapy.
Collapse
Affiliation(s)
- Xiaobin Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
23
|
Lin X, Kang K, Chen P, Zeng Z, Li G, Xiong W, Yi M, Xiang B. Regulatory mechanisms of PD-1/PD-L1 in cancers. Mol Cancer 2024; 23:108. [PMID: 38762484 PMCID: PMC11102195 DOI: 10.1186/s12943-024-02023-w] [Citation(s) in RCA: 130] [Impact Index Per Article: 130.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/10/2024] [Indexed: 05/20/2024] Open
Abstract
Immune evasion contributes to cancer growth and progression. Cancer cells have the ability to activate different immune checkpoint pathways that harbor immunosuppressive functions. The programmed death protein 1 (PD-1) and programmed cell death ligands (PD-Ls) are considered to be the major immune checkpoint molecules. The interaction of PD-1 and PD-L1 negatively regulates adaptive immune response mainly by inhibiting the activity of effector T cells while enhancing the function of immunosuppressive regulatory T cells (Tregs), largely contributing to the maintenance of immune homeostasis that prevents dysregulated immunity and harmful immune responses. However, cancer cells exploit the PD-1/PD-L1 axis to cause immune escape in cancer development and progression. Blockade of PD-1/PD-L1 by neutralizing antibodies restores T cells activity and enhances anti-tumor immunity, achieving remarkable success in cancer therapy. Therefore, the regulatory mechanisms of PD-1/PD-L1 in cancers have attracted an increasing attention. This article aims to provide a comprehensive review of the roles of the PD-1/PD-L1 signaling in human autoimmune diseases and cancers. We summarize all aspects of regulatory mechanisms underlying the expression and activity of PD-1 and PD-L1 in cancers, including genetic, epigenetic, post-transcriptional and post-translational regulatory mechanisms. In addition, we further summarize the progress in clinical research on the antitumor effects of targeting PD-1/PD-L1 antibodies alone and in combination with other therapeutic approaches, providing new strategies for finding new tumor markers and developing combined therapeutic approaches.
Collapse
Affiliation(s)
- Xin Lin
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China
| | - Kuan Kang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China
| | - Pan Chen
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China
| | - Mei Yi
- Department of Dermotology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
- FuRong Laboratory, Changsha, 410078, Hunan, China.
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China.
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
24
|
Javed SA, Najmi A, Ahsan W, Zoghebi K. Targeting PD-1/PD-L-1 immune checkpoint inhibition for cancer immunotherapy: success and challenges. Front Immunol 2024; 15:1383456. [PMID: 38660299 PMCID: PMC11039846 DOI: 10.3389/fimmu.2024.1383456] [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: 02/07/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The programmed death-1 receptor (PD-1) acts as a T-cell brake, and its interaction with ligand-1 (PD-L-1) interferes with signal transduction of the T-cell receptor. This leads to suppression of T-cell survival, proliferation, and activity in the tumor microenvironment resulting in compromised anticancer immunity. PD-1/PD-L-1 interaction blockade shown remarkable clinical success in various cancer immunotherapies. To date, most PD-1/PD-L-1 blockers approved for clinical use are monoclonal antibodies (mAbs); however, their therapeutic use are limited owing to poor clinical responses in a proportion of patients. mAbs also displayed low tumor penetration, steep production costs, and incidences of immune-related side effects. This strongly indicates the importance of developing novel inhibitors as cancer immunotherapeutic agents. Recently, advancements in the small molecule-based inhibitors (SMIs) that directly block the PD-1/PD-L-1 axis gained attention from the scientific community involved in cancer research. SMIs demonstrated certain advantages over mAbs, including longer half-lives, low cost, greater cell penetration, and possibility of oral administration. Currently, several SMIs are in development pipeline as potential therapeutics for cancer immunotherapy. To develop new SMIs, a wide range of structural scaffolds have been explored with excellent outcomes; biphenyl-based scaffolds are most studied. In this review, we analyzed the development of mAbs and SMIs targeting PD-1/PD-L-1 axis for cancer treatment. Altogether, the present review delves into the problems related to mAbs use and a detailed discussion on the development and current status of SMIs. This article may provide a comprehensive guide to medicinal chemists regarding the potential structural scaffolds required for PD-1/PD-L-1 interaction inhibition.
Collapse
Affiliation(s)
| | - Asim Najmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | | | | |
Collapse
|
25
|
Lee JY, Bhandare RR, Boddu SHS, Shaik AB, Saktivel LP, Gupta G, Negi P, Barakat M, Singh SK, Dua K, Chellappan DK. Molecular mechanisms underlying the regulation of tumour suppressor genes in lung cancer. Biomed Pharmacother 2024; 173:116275. [PMID: 38394846 DOI: 10.1016/j.biopha.2024.116275] [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: 11/24/2023] [Revised: 01/30/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Tumour suppressor genes play a cardinal role in the development of a large array of human cancers, including lung cancer, which is one of the most frequently diagnosed cancers worldwide. Therefore, extensive studies have been committed to deciphering the underlying mechanisms of alterations of tumour suppressor genes in governing tumourigenesis, as well as resistance to cancer therapies. In spite of the encouraging clinical outcomes demonstrated by lung cancer patients on initial treatment, the subsequent unresponsiveness to first-line treatments manifested by virtually all the patients is inherently a contentious issue. In light of the aforementioned concerns, this review compiles the current knowledge on the molecular mechanisms of some of the tumour suppressor genes implicated in lung cancer that are either frequently mutated and/or are located on the chromosomal arms having high LOH rates (1p, 3p, 9p, 10q, 13q, and 17p). Our study identifies specific genomic loci prone to LOH, revealing a recurrent pattern in lung cancer cases. These loci, including 3p14.2 (FHIT), 9p21.3 (p16INK4a), 10q23 (PTEN), 17p13 (TP53), exhibit a higher susceptibility to LOH due to environmental factors such as exposure to DNA-damaging agents (carcinogens in cigarette smoke) and genetic factors such as chromosomal instability, genetic mutations, DNA replication errors, and genetic predisposition. Furthermore, this review summarizes the current treatment landscape and advancements for lung cancers, including the challenges and endeavours to overcome it. This review envisages inspired researchers to embark on a journey of discovery to add to the list of what was known in hopes of prompting the development of effective therapeutic strategies for lung cancer.
Collapse
Affiliation(s)
- Jia Yee Lee
- School of Health Sciences, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Richie R Bhandare
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates.
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates
| | - Afzal B Shaik
- St. Mary's College of Pharmacy, St. Mary's Group of Institutions Guntur, Affiliated to Jawaharlal Nehru Technological University Kakinada, Chebrolu, Guntur, Andhra Pradesh 522212, India; Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Lakshmana Prabu Saktivel
- Department of Pharmaceutical Technology, University College of Engineering (BIT Campus), Anna University, Tiruchirappalli 620024, India
| | - Gaurav Gupta
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; School of Pharmacy, Suresh Gyan Vihar University, Jaipur, Rajasthan 302017, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University, PO Box 9, Solan, Himachal Pradesh 173229, India
| | - Muna Barakat
- Department of Clinical Pharmacy & Therapeutics, Applied Science Private University, Amman-11937, Jordan
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara 144411, India; Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney 2007, Australia
| | - Kamal Dua
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
26
|
Donnelly DJ, Kim J, Tran T, Scola PM, Tenney D, Pena A, Petrone T, Zhang Y, Boy KM, Poss MA, Cole EL, Soars MG, Johnson BM, Cohen D, Batalla D, Chow PL, Shorts AO, Du S, Meanwell NA, Bonacorsi SJ. The discovery and evaluation of [ 18F]BMS-986229, a novel macrocyclic peptide PET radioligand for the measurement of PD-L1 expression and in-vivo PD-L1 target engagement. Eur J Nucl Med Mol Imaging 2024; 51:978-990. [PMID: 38049658 DOI: 10.1007/s00259-023-06527-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 11/14/2023] [Indexed: 12/06/2023]
Abstract
PURPOSE A same-day PET imaging agent capable of measuring PD-L1 status in tumors is an important tool for optimizing PD-1 and PD-L1 treatments. Herein we describe the discovery and evaluation of a novel, fluorine-18 labeled macrocyclic peptide-based PET ligand for imaging PD-L1. METHODS [18F]BMS-986229 was synthesized via copper mediated click-chemistry to yield a PD-L1 PET ligand with picomolar affinity and was tested as an in-vivo tool for assessing PD-L1 expression. RESULTS Autoradiography showed an 8:1 binding ratio in L2987 (PD-L1 (+)) vs. HT-29 (PD-L1 (-)) tumor tissues, with >90% specific binding. Specific radioligand binding (>90%) was observed in human non-small-cell lung cancer (NSCLC) and cynomolgus monkey spleen tissues. Images of PD-L1 (+) tissues in primates were characterized by high signal-to-noise, with low background signal in non-expressing tissues. PET imaging enabled clear visualization of PD-L1 expression in a murine model in vivo, with 5-fold higher uptake in L2987 (PD-L1 (+)) than in control HT-29 (PD-L1 (-)) tumors. Moreover, this imaging agent was used to measure target engagement of PD-L1 inhibitors (peptide or mAb), in PD-L1 (+) tumors as high as 97%. CONCLUSION A novel 18F-labeled macrocyclic peptide radioligand was developed for PET imaging of PD-L1 expressing tissues that demonstrated several advantages within a nonhuman primate model when compared directly to adnectin- or mAb-based ligands. Clinical studies are currently evaluating [18F]BMS-986229 to measure PD-L1 expression in tumors.
Collapse
Affiliation(s)
- David J Donnelly
- Small Molecule Drug Discovery-PET Radiochemical Synthesis, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, NJ, 08543, USA.
| | | | - Tritin Tran
- Small Molecule Drug Discovery-PET Radiochemical Synthesis, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, NJ, 08543, USA
| | - Paul M Scola
- Small Molecule Drug Discovery, Bristol Myers Squibb, Cambridge, USA
| | | | | | | | - Yunhui Zhang
- Small Molecule Drug Discovery, Bristol Myers Squibb, Cambridge, USA
| | - Kenneth M Boy
- Small Molecule Drug Discovery, Bristol Myers Squibb, Cambridge, USA
| | - Michael A Poss
- Small Molecule Drug Discovery, Bristol Myers Squibb, Princeton, USA
| | - Erin L Cole
- Small Molecule Drug Discovery-PET Radiochemical Synthesis, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, NJ, 08543, USA
| | - Matthew G Soars
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Cambridge, USA
| | - Benjamin M Johnson
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Cambridge, USA
| | - Daniel Cohen
- Biologics and Platforms, Bristol Myers Squibb, Princeton, USA
| | - Daniel Batalla
- Small Molecule Drug Discovery-PET Radiochemical Synthesis, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, NJ, 08543, USA
| | | | | | - Shuyan Du
- Imaging, Bristol Myers Squibb, Princeton, USA
| | | | - Samuel J Bonacorsi
- Small Molecule Drug Discovery-PET Radiochemical Synthesis, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, NJ, 08543, USA
| |
Collapse
|
27
|
Park W, Han JH, Wei S, Yang ES, Cheon SY, Bae SJ, Ryu D, Chung HS, Ha KT. Natural Product-Based Glycolysis Inhibitors as a Therapeutic Strategy for Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor-Resistant Non-Small Cell Lung Cancer. Int J Mol Sci 2024; 25:807. [PMID: 38255882 PMCID: PMC10815680 DOI: 10.3390/ijms25020807] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide. Targeted therapy against the epidermal growth factor receptor (EGFR) is a promising treatment approach for NSCLC. However, resistance to EGFR tyrosine kinase inhibitors (TKIs) remains a major challenge in its clinical management. EGFR mutation elevates the expression of hypoxia-inducible factor-1 alpha to upregulate the production of glycolytic enzymes, increasing glycolysis and tumor resistance. The inhibition of glycolysis can be a potential strategy for overcoming EGFR-TKI resistance and enhancing the effectiveness of EGFR-TKIs. In this review, we specifically explored the effectiveness of pyruvate dehydrogenase kinase inhibitors and lactate dehydrogenase A inhibitors in combating EGFR-TKI resistance. The aim was to summarize the effects of these natural products in preclinical NSCLC models to provide a comprehensive understanding of the potential therapeutic effects. The study findings suggest that natural products can be promising inhibitors of glycolytic enzymes for the treatment of EGFR-TKI-resistant NSCLC. Further investigations through preclinical and clinical studies are required to validate the efficacy of natural product-based glycolytic inhibitors as innovative therapeutic modalities for NSCLC.
Collapse
Affiliation(s)
- Wonyoung Park
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Jung Ho Han
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Shibo Wei
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Eun-Sun Yang
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Se-Yun Cheon
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Sung-Jin Bae
- Department of Molecular Biology and Immunology, Kosin University College of Medicine, Busan 49267, Republic of Korea;
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea;
| | - Hwan-Suck Chung
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| |
Collapse
|
28
|
Rawat S, Subramaniam K, Subramanian SK, Subbarayan S, Dhanabalan S, Chidambaram SKM, Stalin B, Roy A, Nagaprasad N, Aruna M, Tesfaye JL, Badassa B, Krishnaraj R. Drug Repositioning Using Computer-aided Drug Design (CADD). Curr Pharm Biotechnol 2024; 25:301-312. [PMID: 37605405 DOI: 10.2174/1389201024666230821103601] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/03/2023] [Accepted: 03/20/2023] [Indexed: 08/23/2023]
Abstract
Drug repositioning is a method of using authorized drugs for other unusually complex diseases. Compared to new drug development, this method is fast, low in cost, and effective. Through the use of outstanding bioinformatics tools, such as computer-aided drug design (CADD), computer strategies play a vital role in the re-transformation of drugs. The use of CADD's special strategy for target-based drug reuse is the most promising method, and its realization rate is high. In this review article, we have particularly focused on understanding the various technologies of CADD and the use of computer-aided drug design for target-based drug reuse, taking COVID-19 and cancer as examples. Finally, it is concluded that CADD technology is accelerating the development of repurposed drugs due to its many advantages, and there are many facts to prove that the new ligand-targeting strategy is a beneficial method and that it will gain momentum with the development of technology.
Collapse
Affiliation(s)
- Sona Rawat
- School of Life Sciences, Jaipur National University, Jaipur-302017, India
| | - Kanmani Subramaniam
- Department of Civil Engineering, KPR Institute of Engineering and Technology, Coimbatore-641407, Tamil Nadu, India
| | - Selva Kumar Subramanian
- Department of Sciences, Amrita School of Engineering, Coimbatore - 641112, Tamil Nadu, India
| | - Saravanan Subbarayan
- Department of Civil Engineering, National Institute of Technology, Trichy-620015, Tamil Nadu, India
| | - Subramanian Dhanabalan
- Department of Mechanical Engineering, M. Kumarasamy College of Engineering, Karur - 639113, Tamil Nadu, India
| | | | - Balasubramaniam Stalin
- Department of Mechanical Engineering, Anna University, Regional Campus Madurai, Madurai - 625 019, Tamil Nadu, India
| | - Arpita Roy
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida 201310, India
| | - Nagaraj Nagaprasad
- Department of Mechanical Engineering, ULTRA College of Engineering and Technology, Madurai - 625104, Tamilnadu, India
| | - Mahalingam Aruna
- College of Engineering and Computing, Al Ghurair University, Academic City, Dubai, UAE
| | - Jule Leta Tesfaye
- Dambi Dollo University, College of Natural and Computational Science, Department of Physics, Ethiopia
- Centre for Excellence-Indigenous Knowledge, Innovative Technology Transfer and Entrepreneurship, Dambi Dollo University, Dambi Dollo, Ethiopia
- Ministry of innovation and technology, Ethiopia
| | - Bayissa Badassa
- Department of Mechanical Engineering, Dambi Dollo University, Dambi Dollo, Ethiopia
| | - Ramaswamy Krishnaraj
- Centre for Excellence-Indigenous Knowledge, Innovative Technology Transfer and Entrepreneurship, Dambi Dollo University, Dambi Dollo, Ethiopia
- Ministry of innovation and technology, Ethiopia
- Department of Mechanical Engineering, Dambi Dollo University, Dambi Dollo, Ethiopia
| |
Collapse
|
29
|
Lu Y, Zhang X, Ning J, Zhang M. Immune checkpoint inhibitors as first-line therapy for non-small cell lung cancer: A systematic evaluation and meta-analysis. Hum Vaccin Immunother 2023; 19:2169531. [PMID: 36715018 PMCID: PMC10038046 DOI: 10.1080/21645515.2023.2169531] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 01/04/2023] [Accepted: 01/13/2023] [Indexed: 01/31/2023] Open
Abstract
Recently, immune checkpoint inhibitors (ICIs) present promising application prospects in treating non-small cell lung cancer (NSCLC). This study aimed to investigate optimal treatment strategy by comparing the first-line treatment strategies with ICIs in NSCLC. We retrieved relevant studies on first-line therapy of NSCLC with ICIs. Primary outcomes were overall survival (OS) and progression-free survival (PFS). Secondary outcomes were treatment-related serious adverse events (tr-SAEs) with grade 3 or higher and objective response rate (ORR). We also conducted a Bayesian network meta-analysis. We included 14 studies involving 7,823 patients and compared seven different interventions. In PD-L1 nonselective NSCLC, nivolumab+ipilimumab had good PFS and ORR, pembrolizumab significantly prolonged OS, and nivolumab had the fewest adverse events (AEs). For PD-L1-positive patients, nivolumab remarkably prolonged OS. For those with negative PD-L1, nivolumab+ipilimumab also showed an advantage. In addition, nivolumab+ipilimumab significantly prolonged the PFS in both PD-L1-negative and -positive patients. For patients with PD-L1 tumor proportion score (TPS) within 1-49%, atezolizumab+chemotherapy remarkably prolonged PFS and OS. For those with PD-L1 TPS ≥50%, pembrolizumab prolonged OS and atezolizumab+chemotherapy significantly prolonged PFS. Nivolumab combined with ipilimumab showed advantages in OS, PFS and ORR in most patients. Nivolumab+ipilimumab may be the optimal first-line therapy for NSCLC.
Collapse
Affiliation(s)
- Yu Lu
- Department of General Practice, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiaoyan Zhang
- Department of General Practice, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jiyu Ning
- Department of General Practice, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Manyan Zhang
- Department of Respiration, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
30
|
Larsen TV, Maansson CT, Daugaard TF, Andresen BS, Sorensen BS, Nielsen AL. Trans-Regulation of Alternative PD-L1 mRNA Processing by CDK12 in Non-Small-Cell Lung Cancer Cells. Cells 2023; 12:2844. [PMID: 38132164 PMCID: PMC10741404 DOI: 10.3390/cells12242844] [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: 10/17/2023] [Revised: 11/10/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
Immunotherapy using checkpoint inhibitors targeting the interaction between PD-1 on T cells and PD-L1 on cancer cells has shown significant results in non-small-cell lung cancer (NSCLC). Not all patients respond to the therapy, and PD-L1 expression heterogeneity is proposed to be one determinant for this. The alternative processing of PD-L1 RNA, which depends on an alternative poly-A site in intron 4, generates a shorter mRNA variant (PD-L1v4) encoding soluble PD-L1 (sPD-L1), relative to the canonical PD-L1v1 mRNA encoding membrane-associated PD-L1 (mPD-L1). This study aimed to identify factors influencing the ratio between these two PD-L1 mRNAs in NSCLC cells. First, we verified the existence of the alternative PD-L1 RNA processing in NSCLC cells, and from in silico analyses, we identified a candidate list of regulatory factors. Examining selected candidates showed that CRISPR/Cas9-generated loss-of-function mutations in CDK12 increased the PD-L1v4/PD-L1v1 mRNA ratio and, accordingly, the sPD-L1/mPD-L1 balance. The CDK12/13 inhibitor THZ531 could also increase the PD-L1v4/PD-L1v1 mRNA ratio and impact the PD-L1 transcriptional response to IFN-γ stimulation. The fact that CDK12 regulates PD-L1 transcript variant formation in NSCLC cells is consistent with CDK12's role in promoting transcriptional elongation over intron-located poly-A sites. This study lays the groundwork for clinical investigations to delineate the implications of the CDK12-mediated balancing of sPD-L1 relative to mPD-L1 for immunotherapeutic responses in NSCLC.
Collapse
Affiliation(s)
- Trine V. Larsen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.V.L.); (C.T.M.); (T.F.D.)
| | - Christoffer T. Maansson
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.V.L.); (C.T.M.); (T.F.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
- Department of Clinical Biochemistry, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Tina F. Daugaard
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.V.L.); (C.T.M.); (T.F.D.)
| | - Brage S. Andresen
- Department of Biology and Molecular Biology, Southern University of Denmark, 5230 Odense, Denmark;
| | - Boe S. Sorensen
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
- Department of Clinical Biochemistry, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Anders L. Nielsen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.V.L.); (C.T.M.); (T.F.D.)
| |
Collapse
|
31
|
Wu B, Wang Z, Xu H, Chu X, Jiang Q. NCK1-AS1 promotes the proliferation, migration, invasion, and EMT of non-small cell lung cancer by regulating the miR-361-5p/ADAM10 axis. Histol Histopathol 2023; 38:1453-1464. [PMID: 36912070 DOI: 10.14670/hh-18-604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Lung cancer, one of the most frequently diagnosed cancers, causes a huge number of mortalities globally. Among lung cancers, non-small cell lung cancer (NSCLC) is the most recorded. Despite accumulating research, the molecular basis of NSCLC progression remains poorly known. Therefore, we aim to assess the function of NCK1-AS1 in NSCLC and elucidate the molecular mechanism. Firstly, we quantified the NCK1-AS1 level in tumors and adjacent healthy tissues. NCK1-AS1 was significantly upregulated in NSCLC tumors, which was associated with poor prognosis in patients. Silencing NCK1-AS1 significantly inhibited the proliferation, migration, and invasion, as well as the EMT of NSCLC cell lines. Starbase bioinformatic prediction revealed that NCK1-AS1 targets miR-361-5p which acts to regulate ADAM10 gene expression. Our result showed that NCK1-AS1 upregulation markedly reduced miR-361-5p mRNA expression, while increasing ADAM10 expression. For the first time, we demonstrated that NCK1-AS1 regulates the miR-361-5p/ADAM10 axis, thereby promoting NSCLC progression. NCK1-AS1 might be developed as a therapeutic target for treating NSCLC.
Collapse
Affiliation(s)
- Bingchen Wu
- Department of Oncology, Hospital of Chinese Medicine of Changxing County, Huzhou, Zhejiang, China
| | - Zizong Wang
- Department of Thoracic Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hanlin Xu
- Department of Thoracic Surgery, the Affiliated Hospital of Qingdao, Qingdao, Shandong, China
| | - Xiangyang Chu
- Department of Thoracic Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Qiwen Jiang
- Department of Surgery, Hospital of Chinese Medicine of Changxing County, Huzhou, Zhejiang, China.
| |
Collapse
|
32
|
Wang N, Wang H. Identification of metabolism-related gene signature in lung adenocarcinoma. Medicine (Baltimore) 2023; 102:e36267. [PMID: 38013279 PMCID: PMC10681599 DOI: 10.1097/md.0000000000036267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 11/01/2023] [Indexed: 11/29/2023] Open
Abstract
AIM Lung cancer is one of the most common cancers in China and has a high mortality rate. Most patients who are diagnosed have lost the opportunity to undergo surgery. Aberrant metabolism is closely associated with tumorigenesis. We aimed to identify an effective metabolism-related prediction model for assessing prognosis based on the cancer genome atlas (TCGA) and GSE116959 databases. METHODS TCGA and GSE116959 datasets from Gene Expression Omnibus were used to obtain lung adenocarcinoma (LUAD) data. Additionally, we captured metabolism-related genes (MRGs) from the GeneCards database. First, we extracted differentially expressed genes using R to analyze the LUAD data. We then selected the same differentially expressed genes, including 168 downregulated and 77 upregulated genes. Finally, 218 differentially expressed MRGs (DEMRGs) were included to perform functional enrichment analysis and construct a protein-protein interaction network with the help of Cytoscape and Search Tool for the Retrieval of Interacting Genes database. Cytoscape was used to visualize the intensive intervals in the network. Then univariate and Least Absolute Shrinkage and Selection Operator Cox regression analyses, which assisted in identifying the overall survival (OS)-related DEMRGs and building a 10-DEMRG prognosis model, were performed. The prognostic values, tumor immunity relevance, and molecular mechanism were further investigated. A nomogram incorporating signature, age, gender, and TNM stage was established. RESULTS A 10-DEMRG model was established to forecast the OS of LUAD through Least Absolute Shrinkage and Selection Operator regression analysis. This prognostic signature stratified LUAD patients into low-risk and high-risk groups. The receiver operating characteristic curve and K-M analysis indicated good performance of the DEMRGs signature at predicting OS in the TCGA dataset. Univariate and multivariate Cox regression also revealed that the DEMRGs signature was an independent prognosis factor in LUAD. We noticed that the risk score was substantially related to the clinical parameters of LUAD patients, covering age and stage. Immune analysis results showed that risk score was associated with some immune cells and immune checkpoints. Nomogram also verified the clinical value of the DEMRGs signature. CONCLUSION In this study, we constructed a DEMRGs signature and established a prognostic nomogram that is robust and reliable to predict OS in LUAD. Overall, the findings could help with therapeutic customization and personalized therapies.
Collapse
Affiliation(s)
- Ning Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Hui Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| |
Collapse
|
33
|
Zhao Y, Zhou D, Yuan Y, Chen Y, Zhang K, Tan Y, Fang S. MAPKAPK5-AS1/miR-515-5p/CAB39 Axis Contributes to Non-small Cell Lung Cancer Cell Proliferation and Migration. Mol Biotechnol 2023; 65:1887-1897. [PMID: 36867352 DOI: 10.1007/s12033-023-00654-w] [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: 09/30/2022] [Accepted: 01/04/2023] [Indexed: 03/04/2023]
Abstract
Several studies have elucidated the pivotal function that long noncoding RNAs (lncRNAs) exerted on the initiation and development of various human carcinomas, encompassing non-small cell lung cancer (NSCLC). In spite of the fact that lncRNA MAPKAPK5 antisense RNA 1 (MAPKAPK5-AS1) has already been investigated by researchers and confirmed to play oncogenic roles in colorectal cancer, the underlying regulatory function of MAPKAPK5-AS1 in NSCLC cells still remain unclear. In our research, we found that MAPKAPK5-AS1 was expressed at high levels in NSCLC cells. Biological functional assays unclosed that downregulation of MAPKAPK5-AS1 repressed proliferative and migratory capacities whereas promoted apoptotic level in NSCLC cells. Molecular mechanism experiments confirmed that, in NSCLC cells, MAPKAPK5-AS1 combined with miR-515-5p and negatively modulated miR-515-5p expression level. Besides, calcium-binding protein 39 (CAB39) expression level was verified to be negatively modulated by miR-515-5p whereas positively modulated by MAPKAPK5-AS1 in NSCLC cells. Furthermore, rescued-function assays disclosed that inhibited miR-515-5p expression or overexpressed CAB39 could restore the suppressive influence of MAPKAPK5-AS1 silence on NSCLC progression. In summary, MAPKAPK5-AS1 upregulates CAB39 expression level to facilitate NSCLC progression by sequestering miR-515-5p, providing promising biomarkers for NSCLC treatment.
Collapse
Affiliation(s)
- Yueming Zhao
- Department of Respiratory, Nanjing First Hospital, Qinhuai District, No.68, Changle Road, Nanjing, 210012, Jiangsu, China
| | - Danyang Zhou
- Department of Respiratory, Nanjing First Hospital, Qinhuai District, No.68, Changle Road, Nanjing, 210012, Jiangsu, China
| | - Yuan Yuan
- Department of Respiratory, Nanjing First Hospital, Qinhuai District, No.68, Changle Road, Nanjing, 210012, Jiangsu, China
| | - Yubao Chen
- Department of Respiratory, Nanjing First Hospital, Qinhuai District, No.68, Changle Road, Nanjing, 210012, Jiangsu, China
| | - Kai Zhang
- Department of Respiratory, Nanjing First Hospital, Qinhuai District, No.68, Changle Road, Nanjing, 210012, Jiangsu, China
| | - Yan Tan
- Department of Respiratory, Nanjing First Hospital, Qinhuai District, No.68, Changle Road, Nanjing, 210012, Jiangsu, China.
| | - Surong Fang
- Department of Respiratory, Nanjing First Hospital, Qinhuai District, No.68, Changle Road, Nanjing, 210012, Jiangsu, China.
| |
Collapse
|
34
|
Tian Z, Yang Z, Jin M, Ding R, Wang Y, Chai Y, Wu J, Yang M, Zhao W. Identification of cytokine-predominant immunosuppressive class and prognostic risk signatures in glioma. J Cancer Res Clin Oncol 2023; 149:13185-13200. [PMID: 37479756 DOI: 10.1007/s00432-023-05173-4] [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: 06/11/2023] [Accepted: 07/09/2023] [Indexed: 07/23/2023]
Abstract
PURPOSE The advent of immune checkpoint blockade (ICB) therapies this year has changed the way glioblastoma (GBM) is treated. Meanwhile, some patients with strong PD-L1 expression remain immune checkpoint resistant. To better understand the molecular processes that influence the immune environment, there is an urgent need to characterize the immunosuppressive tumor microenvironment and identify biomarkers to predict patient survival outcomes. PATIENTS AND METHODS Our study analyzed RNA-sequencing data from 178 GBM samples. Their unique gene expression patterns in the tumor microenvironment were analyzed by an unsupervised clustering algorithm. Through these expression patterns, a panel of T-cell exhaustion signatures, immunosuppressive cells, and clinical features correlates with immunotherapy response. The presence or absence of immune status and prognostic signatures was then validated with the test dataset. RESULTS 38.2% of GBM patients showed increased expression of anti-inflammatory cytokines, significant enrichment of T cell exhaustion signals, higher proportion of immunosuppressive cells (macrophages and CD4 regulatory T cells) and nine inhibitory checkpoints (CTLA4, PDCD1, LAG3, BTLA, TIGIT, HAVCR2, IDO1, SIGLEC7, and VISTA). The immunodepleted class (IDC) was used to classify these immunocompromised individuals. Despite the high density of tumor-infiltrating lymphocytes shown by IDC, such patients have a poor prognosis. Although PD-L1 was highly expressed in IDC, it suggested that there might be ICB resistance. There are many IDC predictive signatures to discover. CONCLUSION PD-1 is strongly expressed in a novel immunosuppressive class of GBM, but this cluster may be resistant to ICB therapy. A comprehensive description of this drug-resistant tumor microenvironment could provide new insights into drug resistance mechanisms and improved immunotherapy techniques.
Collapse
Affiliation(s)
- Ziyue Tian
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Zhongyi Yang
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Meng Jin
- The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Ran Ding
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, Guangdong, China
| | - Yuhan Wang
- School of Medical Informatics Engineering, Changchun University of Chinese Medicine, Changchun, 130118, Jilin, China
| | - Yuying Chai
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Jinpu Wu
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Miao Yang
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Weimin Zhao
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
35
|
Malik P, Rani R, Solanki R, Patel VH, Mukherjee TK. Understanding the feasibility of chemotherapeutic and immunotherapeutic targets against non-small cell lung cancers: an update of resistant responses and recent combinatorial therapies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:850-895. [PMID: 37970206 PMCID: PMC10645466 DOI: 10.37349/etat.2023.00171] [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: 02/26/2023] [Accepted: 05/17/2023] [Indexed: 11/17/2023] Open
Abstract
Despite consistent progress in prompt diagnosis and curative therapies in the last decade, lung cancer (LC) continues to threaten mankind, accounting for nearly twice the casualties compared to prostate, breast, and other cancers. Statistics associate ~25% of 2021 cancer-related deaths with LC, more than 80% of which are explicitly caused by tobacco smoking. Prevailing as small and non-small cell pathologies, with respective occurring frequency of nearly 15% and 80-85%, non-small cell LCs (NSCLCs) are prominently distinguished into lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), subtypes. Since the first use of epidermal growth factor receptor (EGFR) inhibitor gefitinib for NSCLC treatment in 2002, immense progress has been made for targeted therapies with the next generation of drugs spanning across the chronological generations of small molecule inhibitors. The last two years have overseen the clinical approval of more than 10 therapeutic agents as first-line NSCLC medications. However, uncertain mutational aberrations as well as systemic resistant responses, and abysmal overall survival curtail the combating efficacies. Of late, immune checkpoint inhibitors (ICIs) against various molecules including programmed cell death-1 (PD-1) and its ligand (PD-L1) have been demonstrated as reliable LC treatment targets. Keeping these aspects in mind, this review article discusses the success of NSCLC chemo and immunotherapies with their characteristic effectiveness and future perspectives.
Collapse
Affiliation(s)
- Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Ruma Rani
- Indian Council of Agricultural Research (ICAR)-National Research Centre on Equines, Hisar 125001, Haryana, India
| | - Raghu Solanki
- School of Life Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | | | | |
Collapse
|
36
|
Sooi K, Walsh R, Kumarakulasinghe N, Wong A, Ngoi N. A review of strategies to overcome immune resistance in the treatment of advanced prostate cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:656-673. [PMID: 37842236 PMCID: PMC10571060 DOI: 10.20517/cdr.2023.48] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/06/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023]
Abstract
Immunotherapy has become integral in cancer therapeutics over the past two decades and is now part of standard-of-care treatment in multiple cancer types. While various biomarkers and pathway alterations such as dMMR, CDK12, and AR-V7 have been identified in advanced prostate cancer to predict immunotherapy responsiveness, the vast majority of prostate cancer remain intrinsically immune-resistant, as evidenced by low response rates to anti-PD(L)1 monotherapy. Since regulatory approval of the vaccine therapy sipuleucel-T in the biomarker-unselected population, there has not been much success with immunotherapy treatment in advanced prostate cancer. Researchers have looked at various strategies to overcome immune resistance, including the identification of more biomarkers and the combination of immunotherapy with existing effective prostate cancer treatments. On the horizon, novel drugs using bispecific T-cell engager (BiTE) and chimeric antigen receptors (CAR) technology are being explored and have shown promising early efficacy in this disease. Here we discuss the features of the tumour microenvironment that predispose to immune resistance and rational strategies to enhance antitumour responsiveness in advanced prostate cancer.
Collapse
Affiliation(s)
| | | | | | | | - Natalie Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore 119228, Singapore
| |
Collapse
|
37
|
Bienkowska KJ, Hanley CJ. Improved understanding of NSCLC immunotherapy response mechanisms from single-cell analysis. Transl Lung Cancer Res 2023; 12:1807-1811. [PMID: 37691874 PMCID: PMC10483077 DOI: 10.21037/tlcr-23-428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/01/2023] [Indexed: 09/12/2023]
|
38
|
Seo H, Verma A, Kinzel M, Huang Q, Mahoney DJ, Jacquelot N. Targeting Potential of Innate Lymphoid Cells in Melanoma and Other Cancers. Pharmaceutics 2023; 15:2001. [PMID: 37514187 PMCID: PMC10384206 DOI: 10.3390/pharmaceutics15072001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Reinvigorating the killing function of tumor-infiltrating immune cells through the targeting of regulatory molecules expressed on lymphocytes has markedly improved the prognosis of cancer patients, particularly in melanoma. While initially thought to solely strengthen adaptive T lymphocyte anti-tumor activity, recent investigations suggest that other immune cell subsets, particularly tissue-resident innate lymphoid cells (ILCs), may benefit from immunotherapy treatment. Here, we describe the recent findings showing immune checkpoint expression on tissue-resident and tumor-infiltrating ILCs and how their effector function is modulated by checkpoint blockade-based therapies in cancer. We discuss the therapeutic potential of ILCs beyond the classical PD-1 and CTLA-4 regulatory molecules, exploring other possibilities to manipulate ILC effector function to further impede tumor growth and quench disease progression.
Collapse
Affiliation(s)
- Hobin Seo
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Amisha Verma
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Megan Kinzel
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Qiutong Huang
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, QLD 4102, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Douglas J Mahoney
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Nicolas Jacquelot
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
39
|
Ahn J, Nagasaka M. Spotlight on Cemiplimab-rwlc in the Treatment of Non-Small Cell Lung Cancer (NSCLC): Focus on Patient Selection and Considerations. Cancer Manag Res 2023; 15:627-634. [PMID: 37457376 PMCID: PMC10349595 DOI: 10.2147/cmar.s325856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
In metastatic non-small cell lung cancer (NSCLC), tumors that do not harbor driver mutations in EGFR or gene fusions in ALK and ROS, PD-1 and PD-L1 inhibitors have become a cornerstone in first line treatment, either as monotherapy or in combination with chemotherapy. This paper reviews cemiplimab-rwlc, the third PD-1/L1 inhibitor to be approved in the setting for first line treatment in NSCLC, as monotherapy or in combination therapy with chemotherapy, to provide a perspective on the subtle differences in patient population for the cemiplimab studies and consideration of its primary and subgroup results in the context of first line therapies for NSCLC.
Collapse
Affiliation(s)
- Jeffrey Ahn
- Department of Medicine, University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange, CA, USA
| | - Misako Nagasaka
- Department of Medicine, University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange, CA, USA
- St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
40
|
Ding R, Wang Y, Fan J, Tian Z, Wang S, Qin X, Su W, Wang Y. Identification of immunosuppressive signature subtypes and prognostic risk signatures in triple-negative breast cancer. Front Oncol 2023; 13:1108472. [PMID: 37377907 PMCID: PMC10292819 DOI: 10.3389/fonc.2023.1108472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 04/10/2023] [Indexed: 06/29/2023] Open
Abstract
Purpose Immune checkpoint blockade (ICB) therapy has transformed the treatment of triple-negative breast cancer (TNBC) in recent years. However, some TNBC patients with high programmed death-ligand 1 (PD-L1) expression levels develop immune checkpoint resistance. Hence, there is an urgent need to characterize the immunosuppressive tumor microenvironment and identify biomarkers to construct prognostic models of patient survival outcomes in order to understand biological mechanisms operating within the tumor microenvironment. Patients and methods RNA sequence (RNA-seq) data from 303 TNBC samples were analyzed using an unsupervised cluster analysis approach to reveal distinctive cellular gene expression patterns within the TNBC tumor microenvironment (TME). A panel of T cell exhaustion signatures, immunosuppressive cell subtypes and clinical features were correlated with the immunotherapeutic response, as assessed according to gene expression patterns. The test dataset was then used to confirm the occurrence of immune depletion status and prognostic features and to formulate clinical treatment recommendations. Concurrently, a reliable risk prediction model and clinical treatment strategy were proposed based on TME immunosuppressive signature differences between TNBC patients with good versus poor survival status and other clinical prognostic factors. Results Significantly enriched TNBC microenvironment T cell depletion signatures were detected in the analyzed RNA-seq data. A high proportion of certain immunosuppressive cell subtypes, 9 inhibitory checkpoints and enhanced anti-inflammatory cytokine expression profiles were noted in 21.4% of TNBC patients that led to the designation of this group of immunosuppressed patients as the immune depletion class (IDC). Although IDC group TNBC samples contained tumor-infiltrating lymphocytes present at high densities, IDC patient prognosis was poor. Notably, PD-L1 expression was relatively elevated in IDC patients that indicated their cancers were resistant to ICB treatment. Based on these findings, a set of gene expression signatures predicting IDC group PD-L1 resistance was identified then used to develop risk models for use in predicting clinical therapeutic outcomes. Conclusion A novel TNBC immunosuppressive tumor microenvironment subtype associated with strong PD-L1 expression and possible resistance to ICB treatment was identified. This comprehensive gene expression pattern may provide fresh insights into drug resistance mechanisms for use in optimizing immunotherapeutic approaches for TNBC patients.
Collapse
Affiliation(s)
- Ran Ding
- Changchun University of Chinese Medicine, Changchun, Jilin, China
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yuhan Wang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jinyan Fan
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ziyue Tian
- The Affiliated Hospital of Changchun University of Chinese Medicine, Jilin, China
| | - Shuang Wang
- Department of Traditional Chinese Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiujuan Qin
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Wei Su
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yanbo Wang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
41
|
Wang Y, Huang S, Feng X, Xu W, Luo R, Zhu Z, Zeng Q, He Z. Advances in efficacy prediction and monitoring of neoadjuvant immunotherapy for non-small cell lung cancer. Front Oncol 2023; 13:1145128. [PMID: 37265800 PMCID: PMC10229830 DOI: 10.3389/fonc.2023.1145128] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) has become mainstream in the treatment of non-small cell lung cancer (NSCLC). The idea of harnessing the immune system to fight cancer is fast developing. Neoadjuvant treatment in NSCLC is undergoing unprecedented change. Chemo-immunotherapy combinations not only seem to achieve population-wide treating coverage irrespective of PD-L1 expression but also enable achieving a pathological complete response (pCR). Despite these recent advancements in neoadjuvant chemo-immunotherapy, not all patients respond favorably to treatment with ICIs plus chemo and may even suffer from severe immune-related adverse effects (irAEs). Similar to selection for target therapy, identifying patients most likely to benefit from chemo-immunotherapy may be valuable. Recently, several prognostic and predictive factors associated with the efficacy of neoadjuvant immunotherapy in NSCLC, such as tumor-intrinsic biomarkers, tumor microenvironment biomarkers, liquid biopsies, microbiota, metabolic profiles, and clinical characteristics, have been described. However, a specific and sensitive biomarker remains to be identified. Recently, the construction of prediction models for ICI therapy using novel tools, such as multi-omics factors, proteomic tests, host immune classifiers, and machine learning algorithms, has gained attention. In this review, we provide a comprehensive overview of the different positive prognostic and predictive factors in treating preoperative patients with ICIs, highlight the recent advances made in the efficacy prediction of neoadjuvant immunotherapy, and provide an outlook for joint predictors.
Collapse
Affiliation(s)
- Yunzhen Wang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sha Huang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangwei Feng
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wangjue Xu
- Department of Thoracic Surgery, Longyou County People’s Hospital, Longyou, China
| | - Raojun Luo
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziyi Zhu
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingxin Zeng
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengfu He
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
42
|
Iturrioz-Rodríguez N, Sampron N, Matheu A. Current advances in temozolomide encapsulation for the enhancement of glioblastoma treatment. Theranostics 2023; 13:2734-2756. [PMID: 37284445 PMCID: PMC10240814 DOI: 10.7150/thno.82005] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/20/2023] [Indexed: 06/08/2023] Open
Abstract
Glioblastoma is the most common and lethal brain tumor in adults. The incorporation of temozolomide (TMZ) into the standard treatment has increased the overall survival rate of glioblastoma patients. Since then, significant advances have been made in understanding the benefits and limitations of TMZ. Among the latter, the unspecific toxicity of TMZ, poor solubility, and hydrolyzation are intrinsic characteristics, whereas the presence of the blood-brain barrier and some tumor properties, such as molecular and cellular heterogeneity and therapy resistance, have limited the therapeutic effects of TMZ in treating glioblastoma. Several reports have revealed that different strategies for TMZ encapsulation in nanocarriers overcome those limitations and have shown that they increase TMZ stability, half-life, biodistribution, and efficacy, offering the promise for future nanomedicine therapies in handling glioblastoma. In this review, we analyze the different nanomaterials used for the encapsulation of TMZ to improve its stability, blood half-life and efficacy, paying special attention to polymer- and lipid-based nanosystems. To improve TMZ drug resistance, present in up to 50% of patients, we detail TMZ combined therapeutic with i) other chemotherapies, ii) inhibitors, iii) nucleic acids, iv) photosensitizers and other nanomaterials for photodynamic therapy, photothermal therapy, and magnetic hyperthermia, v) immunotherapy, and vi) other less explored molecules. Moreover, we describe targeting strategies, such as passive targeting, active targeting to BBB endothelial cells, glioma cells, and glioma cancer stem cells, and local delivery, where TMZ has demonstrated an improved outcome. To finish our study, we include possible future research directions that could help decrease the time needed to move from bench to bedside.
Collapse
Affiliation(s)
| | - Nicolas Sampron
- Cellular Oncology group, Biodonostia Health Research Institute, San Sebastian, Spain
| | - Ander Matheu
- Cellular Oncology group, Biodonostia Health Research Institute, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento (CIBERfes), Carlos III Institute, Madrid, Spain
| |
Collapse
|
43
|
Peng L, Xu Q, Yin S, Zhang Y, Wu H, Liu Y, Chen L, Hu Y, Yuan J, Peng K, Lin Q. The emerging nanomedicine-based technology for non-small cell lung cancer immunotherapy: how far are we from an effective treatment. Front Oncol 2023; 13:1153319. [PMID: 37182180 PMCID: PMC10172578 DOI: 10.3389/fonc.2023.1153319] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a prominent etiology of cancer-related mortality. The heterogeneous nature of this disease impedes its accurate diagnosis and efficacious treatment. Consequently, constant advancements in research are imperative in order to comprehend its intricate nature. In addition to currently available therapies, the utilization of nanotechnology presents an opportunity to enhance the clinical outcomes of NSCLC patients. Notably, the burgeoning knowledge of the interaction between the immune system and cancer itself paves the way for developing novel, emerging immunotherapies for treating NSCLC in the early stages of the disease. It is believed that with the novel engineering avenues of nanomedicine, there is a possibility to overcome the inherent limitations derived from conventional and emerging treatments, such as off-site drug cytotoxicity, drug resistance, and administration methods. Combining nanotechnology with the convergence points of current therapies could open up new avenues for meeting the unmet needs of NSCLC treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Qin Lin
- Department of Thoracic Surgery, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
44
|
Dehghani T, Shahrjerdi A, Kahrizi MS, Soleimani E, Ravandeh S, Merza MS, Rahnama N, Ebrahimzadeh F, Bakhshesh M. Targeting programmed cell death protein 1 (PD-1) for treatment of non-small-cell lung carcinoma (NSCLC); the recent advances. Pathol Res Pract 2023; 246:154470. [PMID: 37150133 DOI: 10.1016/j.prp.2023.154470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/09/2023]
Abstract
The immune system uses various immune checkpoint axes to adjust responses, support homeostasis, and deter self-reactivity and autoimmunity. Nevertheless, non-small-cell lung carcinoma (NSCLC) can use protective mechanisms to facilitate immune evasion, which leads to potentiated cancer survival and proliferation. In this light, many blocking anti-bodies have been developed to negatively regulate checkpoint molecules, in particular, programmed cell death protein 1 (PD-1) / PD-ligand 1 (L1), and bypass these immune suppressive mechanisms. Meanwhile, anti-PD-1 anti-bodies such as nivolumab, pembrolizumab, cemiplimab, and sintilimab have shown excellent competence in successfully inspiring immune responses versus NSCLC. Accordingly, the United States Food and Drug Administration (FDA) has recently approved nivolumab (alone or in combination with ipilimumab) and pembrolizumab (alone or in combination with chemotherapy) as first-line treatment for advanced NSCLC patients. However, PD-1 blockade monotherapy remains inefficient in more than 60% of NSCLC patients, and many patients don't respond or acquire resistance to this modality. Also, toxicities related to anti-PD-1 anti-body have been progressively identified in clinical trials and oncology practice. Herein, we will outline the clinical benefits of PD-1 blockade therapy alone or in combination with other treatments (e.g., chemotherapy, radiotherapy, anti-angiogenic therapy) in NSCLC patients. Moreover, we will take a glimpse into the recently identified predictive biomarkers to determine patients most likely to suffer serious adverse events to decrease untoward toxicity risk and diminish treatment costs.
Collapse
Affiliation(s)
- Tannaz Dehghani
- Department of Internal Medicine, Lorestan University of Medical Sciences, Lorestan, Iran
| | - Alireza Shahrjerdi
- National Institute for Genetic Engineering and Biotechnology (NIGEB), P.O. Box: 14965/161, Tehran, Iran
| | | | - Elnaz Soleimani
- Departmant of Genetic, Babol University of Medical Science, Babol, Iran
| | | | - Muna S Merza
- Prosthetic Dental Techniques Department, Al-Mustaqbal university College, Babylon 51001, Iraq
| | - Negin Rahnama
- Department of Internal Medicine and Health Services, Semnan University of Medical Sciences, Semnan, Iran
| | - Farnoosh Ebrahimzadeh
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Morteza Bakhshesh
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran.
| |
Collapse
|
45
|
Zhang C, Zhang C, Wang H. Immune-checkpoint inhibitor resistance in cancer treatment: Current progress and future directions. Cancer Lett 2023; 562:216182. [PMID: 37076040 DOI: 10.1016/j.canlet.2023.216182] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
Cancer treatment has been advanced with the advent of immune checkpoint inhibitors (ICIs) exemplified by anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), anti-programmed cell death protein 1 (PD-1) and programmed cell death ligand 1 (PD-L1) drugs. Patients have reaped substantial benefit from ICIs in many cancer types. However, few patients benefit from ICIs whereas the vast majority undergoing these treatments do not obtain survival benefit. Even for patients with initial responses, they may encounter drug resistance in their subsequent treatments, which limits the efficacy of ICIs. Therefore, a deepening understanding of drug resistance is critically important for the explorations of approaches to reverse drug resistance and to boost ICI efficacy. In the present review, different mechanisms of ICI resistance have been summarized according to the tumor intrinsic, tumor microenvironment (TME) and host classifications. We further elaborated corresponding strategies to battle against such resistance accordingly, which include targeting defects in antigen presentation, dysregulated interferon-γ (IFN-γ) signaling, neoantigen depletion, upregulation of other T cell checkpoints as well as immunosuppression and exclusion mediated by TME. Moreover, regarding the host, several additional approaches that interfere with diet and gut microbiome have also been described in reversing ICI resistance. Additionally, we provide an overall glimpse into the ongoing clinical trials that utilize these mechanisms to overcome ICI resistance. Finally, we summarize the challenges and opportunities that needs to be addressed in the investigation of ICI resistance mechanisms, with the aim to benefit more patients with cancer.
Collapse
Affiliation(s)
- Chenyue Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Chenxing Zhang
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
46
|
Vardas V, Tolios A, Christopoulou A, Georgoulias V, Xagara A, Koinis F, Kotsakis A, Kallergi G. Immune Checkpoint and EMT-Related Molecules in Circulating Tumor Cells (CTCs) from Triple Negative Breast Cancer Patients and Their Clinical Impact. Cancers (Basel) 2023; 15:1974. [PMID: 37046635 PMCID: PMC10093450 DOI: 10.3390/cancers15071974] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Triple negative breast cancer (TNBC) is the most aggressive breast cancer subtype. There are few targeted therapies for these patients, leading to an unmet need for new biomarkers. The present study aimed to investigate the expression of PD-L1, CTLA-4, GLU, and VIM in CTCs of TNBC patients. Ninety-five patients were enrolled in this study: sixty-four TNBC and thirty-one luminal. Of these patients, 60 were in the early stage, while 35 had metastatic disease. Protein expression was identified by immunofluorescence staining experiments and VyCAP analysis. All the examined proteins were upregulated in TNBC patients. The expression of the GLU+VIM+CK+ phenotype was higher (50%) in metastatic TNBC compared to early TNBC patients (17%) (p = 0.005). Among all the BC patients, a significant correlation was found between PD-L1+CD45-CK+ and CTLA-4+CD45-CK+ phenotypes (Spearman test, p = 0.024), implying an important role of dual inhibition in BC. Finally, the phenotypes GLU+VIM+CK+ and PD-L1+CD45-CK+ were associated with shorter OS in TNBC patients (OS: log-rank p = 0.048, HR = 2.9, OS: log-rank p < 0.001, HR = 8.7, respectively). Thus, PD-L1, CTLA-4, GLU, and VIM constitute significant biomarkers in TNBC associated with patients' outcome, providing new therapeutic targets for this difficult breast cancer subtype.
Collapse
Affiliation(s)
- Vasileios Vardas
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece
| | - Anastasios Tolios
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece
| | | | | | - Anastasia Xagara
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece
| | - Filippos Koinis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Athanasios Kotsakis
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, GR-41110 Larissa, Greece
- Department of Medical Oncology, University General Hospital of Larissa, GR-41110 Larissa, Greece
| | - Galatea Kallergi
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, GR-26504 Patras, Greece
| |
Collapse
|
47
|
Larsen TV, Dybdal N, Daugaard TF, Lade-Keller J, Lin L, Sorensen BS, Nielsen AL. Examination of the Functional Relationship between PD-L1 DNA Methylation and mRNA Expression in Non-Small-Cell Lung Cancer. Cancers (Basel) 2023; 15:cancers15061909. [PMID: 36980795 PMCID: PMC10047551 DOI: 10.3390/cancers15061909] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/04/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Immunotherapy targeting the interaction between programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) is a treatment option for patients with non-small-cell lung cancer (NSCLC). The expression of PD-L1 by the NSCLC cells determines treatment effectiveness, but the relationship between PD-L1 DNA methylation and expression has not been clearly described. We investigated PD-L1 DNA methylation, mRNA expression, and protein expression in NSCLC cell lines and tumor biopsies. We used clustered regularly interspaced short palindromic repeats-associated protein 9 (CRISPR-Cas9) to modify PD-L1 genetic contexts and endonuclease deficient Cas9 (dCas9) fusions with ten-eleven translocation methylcytosine dioxygenase 1 (TET1) and DNA (cytosine-5)-methyltransferase 3A (DNMT3A) to manipulate PD-L1 DNA methylation. In NSCLC cell lines, we identified specific PD-L1 CpG sites with methylation levels inversely correlated with PD-L1 mRNA expression. However, inducing PD-L1 mRNA expression with interferon-γ did not decrease the methylation level for these CpG sites, and using CRISPR-Cas9, we found that the CpG sites did not directly confer a negative regulation. dCas9-TET1 and dCas9-DNMT3A could induce PD-L1 hypo- and hyper-methylation, respectively, with the latter conferring a decrease in expression showing the functional impact of methylation. In NSCLC biopsies, the inverse correlation between the methylation and expression of PD-L1 was weak. We conclude that there is a regulatory link between PD-L1 DNA methylation and expression. However, since these measures are weakly associated, this study highlights the need for further research before PD-L1 DNA methylation can be implemented as a biomarker and drug target for measures to improve the effectiveness of PD-1/PD-L1 immunotherapy in NSCLC.
Collapse
Affiliation(s)
- Trine V Larsen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Nina Dybdal
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Tina F Daugaard
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | | | - Lin Lin
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Boe S Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, 8200 Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Anders L Nielsen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
48
|
Mortezaee K, Majidpoor J. Anti-PD-(L)1 therapy of non-small cell lung cancer-A summary of clinical trials and current progresses. Heliyon 2023; 9:e14566. [PMID: 36950599 PMCID: PMC10025922 DOI: 10.1016/j.heliyon.2023.e14566] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Background This review discusses the impact of mono or combination therapy of immune checkpoint inhibitor (ICI) therapy in non-small cell lung cancer (NSCLC) patients, comparing clinical outcomes and safety. Cancer subtype, tumor mutational burden (TMB), programmed death-ligand 1 (PD-L1) expression state and T cell infiltration (TIL) density are considered for interpretations. Besides, current progresses in the field of immunotherapy are discussed. Results Anti-PD-(L)1 is a safe and an effective strategy in patients with advanced/metastatic NSCLC. Clinical responses to nivolumab and pembrolizumab, in particular, are promising. The most desired clinical responses are for patients receiving combination of anti-PD-(L)1 or anti-PD-(L)1/anti-cytotoxic T lymphocyte associated antigen-4 (CTLA-4) with chemotherapy (taxane and platinum). PD-L1 expression state (PD-L1 ≥ 50%), patient performance state (PS: 0-1 ECOG scale) and effector T cell (Teff) immune signature considerably affect ICI responses. Higher ICI responses are also expected in TMB high but EGFR-/ALK- cancer patients. In regard with safety profile, adverse events (AEs) related to anti-PD-(L)1 are lower compared with that for platinum-based and docetaxel therapy. Toripalimab is the safest among various immunotherapy drugs. Bispecific antibodies against anti-PD-(L)1 with dominant signaling or alternative checkpoints in tumor microenvironment (TME) is the current focus in immunotherapy of cancers like NSCLC. Besides, the contribution of extracellular vesicles (EVs) to immune escape and their implication in cancer diagnosis and therapy is on the eye of current investigations. Conclusion Appropriate biomarker selection will improve therapy outcomes in ICI treated NSCLC patients, particularly in cases under combinatory ICI therapy. Application of bispecific antibodies and EV-based targeted therapy are effective novel strategies to improve therapeutic outcomes in cancer patients.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Corresponding author.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
49
|
Blakely CM, Weder W, Bubendorf L, He J, Majem M, Shyr Y, Chaft JE. Primary endpoints to assess the efficacy of novel therapeutic approaches in epidermal growth factor receptor-mutated, surgically resectable non-small cell lung cancer: A review. Lung Cancer 2023; 177:59-72. [PMID: 36736076 DOI: 10.1016/j.lungcan.2023.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/20/2022] [Accepted: 01/01/2023] [Indexed: 01/04/2023]
Abstract
While the discovery of oncogenic driver mutations has personalized the metastatic non-small cell lung cancer (NSCLC) treatment landscape with effective targeted therapies, implementation of new treatments in resectable NSCLC has been limited due to the long follow-up needed for overall survival (OS). Until recently, treatment for patients with early-stage resectable NSCLC has been limited to perioperative chemotherapy, which provides modest benefits. However, the regulatory acceptance of two surrogate endpoints for OS has allowed recent approval of both adjuvant osimertinib and atezolizumab, providing patients with new treatment options to improve outcomes. In phase 3 oncology trials, OS has historically been viewed as the gold-standard efficacy measure, but disease-free survival and event-free survival (EFS) are now validated surrogate endpoints for OS in clinical trials and should be considered when mature OS data is unavailable. Another potential surrogate endpoint in the adjuvant NSCLC setting is circulating tumor DNA (ctDNA)-based minimal residual disease (MRD), although prospective validation is needed. For neoadjuvant targeted therapies, EFS, major pathologic response and ctDNA-based MRD are potential surrogate endpoints. To fully translate the success of the personalized treatment advances in the metastatic setting to earlier-stage disease, prospective validation studies of these potential surrogate endpoints that can accelerate the evaluation of drug efficacy are needed. A collaborative effort is also needed from all clinical and regulatory parties to collate surrogate endpoint data for large-scale validation. In this review we discuss the trends in surrogate endpoints used in oncology trials, with a focus on considerations for selecting appropriate primary endpoints in early-stage resectable EGFR-mutant NSCLC, an area of unmet need for novel treatment options.
Collapse
Affiliation(s)
- Collin M Blakely
- Department of Medicine and Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Walter Weder
- Department of Thoracic Surgery, University of Zurich (director Emeritus), Thoraxchirurgie, Klinik Bethanien, Zürich, Switzerland
| | - Lukas Bubendorf
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Jianxing He
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jamie E Chaft
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY 10021, USA.
| |
Collapse
|
50
|
Rocco D, Della Gravara L, Battiloro C, Palazzolo G, Gridelli C. Recently approved and emerging monoclonal antibody immune checkpoint inhibitors for the treatment of advanced non-small cell lung cancer. Expert Opin Biol Ther 2023; 23:261-268. [PMID: 36803090 DOI: 10.1080/14712598.2023.2183116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
INTRODUCTION CTLA-4/PD-1/PD-L1- directed immune checkpoint inhibitors (ICIs) are one of the standard therapies for the treatment of advanced non-small cell lung cancer (NSCLC). However, some new classes of monoclonal antibodies are emerging as promising therapies for advanced NSCLC. AREAS COVERED Therefore, this paper aims to provide a comprehensive review of the recently approved as well as emerging monoclonal antibody immune checkpoint inhibitors for the treatment of advanced NSCLC. EXPERT OPINION Further and larger studies will be needed to explore the promising emerging data on new ICIs. Future phase III trials could allow us to properly assess the role of each immune checkpoints in the wider context of the tumor microenvironment and thus the best new ICIs to use, the best approach and the most effective subset of patients to select.
Collapse
Affiliation(s)
- Danilo Rocco
- Department of Pulmonary Oncology, AORN dei Colli Monaldi, Naples, Italy
| | - Luigi Della Gravara
- Department of Precision Medicine, Università degli studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Ciro Battiloro
- Department of Pulmonary Oncology, AORN dei Colli Monaldi, Naples, Italy
| | | | - Cesare Gridelli
- Division of Medical Oncology, S.G. Moscati Hospital, Avellino, Italy
| |
Collapse
|