1
|
Zhang Y, Yang J, Shao T, Chen J, Shu Q, Shou L. Exploration of genetic characterization in hyperprogressive disease after immunotherapy retreatment in a patient with LCNEC: A case report. Hum Vaccin Immunother 2024; 20:2313281. [PMID: 38348622 PMCID: PMC10865920 DOI: 10.1080/21645515.2024.2313281] [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: 09/12/2023] [Accepted: 01/30/2024] [Indexed: 02/15/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have emerged as a promising therapeutic option for large cell neuroendocrine carcinoma (LCNEC). However, various studies have suggested a potential risk of hyperprogressive disease (HPD) in patients receiving ICI, which might be associated with gene alterations. Here, this is the first report on an unknown primary LCNEC patient who had achieved a long-term response from ICI treatment (atezolizumab), but developed HPD after tumor progression due to receiving another ICI agent (serplulimab). The mutation region of FAT4, SMARCA4, CYLD, CTNNB1, and KIT was altered prior to serplulimab treatment compared to before atezolizumab treatment. This case suggested a potential association between these mutated genes and HPD. Patients with the aforementioned genes should caution when selecting ICI treatment. These findings required further confirmation in a larger study cohort.
Collapse
Affiliation(s)
- Yao Zhang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiayao Yang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tianyu Shao
- Department of Oncology, Guang’ Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jialu Chen
- Department of Oncology, Hangzhou Third People’s Hospital, Hangzhou, China
| | - Qijin Shu
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Liumei Shou
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
2
|
Jiang J, Ye P, Sun N, Zhu W, Yang M, Yu M, Yu J, Zhang H, Gao Z, Zhang N, Guo S, Ji Y, Li S, Zhang C, Miao S, Chai M, Liu W, An Y, Hong J, Wei W, Zhang S, Qiu H. Yap methylation-induced FGL1 expression suppresses anti-tumor immunity and promotes tumor progression in KRAS-driven lung adenocarcinoma. Cancer Commun (Lond) 2024. [PMID: 39340215 DOI: 10.1002/cac2.12609] [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: 12/15/2023] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Despite significant strides in lung cancer immunotherapy, the response rates for Kirsten rat sarcoma viral oncogene homolog (KRAS)-driven lung adenocarcinoma (LUAD) patients remain limited. Fibrinogen-like protein 1 (FGL1) is a newly identified immune checkpoint target, and the study of related resistance mechanisms is crucial for improving the treatment outcomes of LUAD patients. This study aimed to elucidate the potential mechanism by which FGL1 regulates the tumor microenvironment in KRAS-mutated cancer. METHODS The expression levels of FGL1 and SET1 histone methyltransferase (SET1A) in lung cancer were assessed using public databases and clinical samples. Lentiviruses were constructed for transduction to overexpress or silence FGL1 in lung cancer cells and mouse models. The effects of FGL1 and Yes-associated protein (Yap) on the immunoreactivity of cytotoxic T cells in tumor tissues were evaluated using immunofluorescence staining and flow cytometry. Chromatin immunoprecipitation and dual luciferase reporter assays were used to study the SET1A-directed transcriptional program. RESULTS Upregulation of FGL1 expression in KRAS-mutated cancer was inversely correlated with the infiltration of CD8+ T cells. Mechanistically, KRAS activated extracellular signal-regulated kinase 1/2 (ERK1/2), which subsequently phosphorylated SET1A and increased its stability and nuclear localization. SET1A-mediated methylation of Yap led to Yap sequestration in the nucleus, thereby promoting Yap-induced transcription of FGL1 and immune evasion in KRAS-driven LUAD. Notably, dual blockade of programmed cell death-1 (PD-1) and FGL1 further increased the therapeutic efficacy of anti-PD-1 immunotherapy in LUAD patients. CONCLUSION FGL1 could be used as a diagnostic biomarker of KRAS-mutated lung cancer, and targeting the Yap-FGL1 axis could increase the efficacy of anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Ji Jiang
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Pengfei Ye
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Ningning Sun
- School of Nursing, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Weihua Zhu
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Mei Yang
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Manman Yu
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Jingjing Yu
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Hui Zhang
- School of Nursing, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Zijie Gao
- School of Nursing, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Ningjie Zhang
- School of Nursing, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Shijie Guo
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Yuru Ji
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Siqi Li
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Cuncun Zhang
- School of Nursing, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Sainan Miao
- School of Nursing, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Mengqi Chai
- School of Nursing, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Wenmin Liu
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Yue An
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Jian Hong
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P. R. China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Shihao Zhang
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, P. R. China
| | - Huan Qiu
- School of Nursing, Anhui Medical University, Hefei, Anhui, P. R. China
| |
Collapse
|
3
|
Pei B, Zhang J, Lai L, Chen H. Neutrophil-to-lymphocyte ratio as a predictive biomarker for hyperprogressive disease mediated by immune checkpoint inhibitors: a systematic review and meta-analysis. Front Immunol 2024; 15:1393925. [PMID: 39380991 PMCID: PMC11460549 DOI: 10.3389/fimmu.2024.1393925] [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: 02/29/2024] [Accepted: 08/27/2024] [Indexed: 10/10/2024] Open
Abstract
Background Hyperprogressive disease (HPD) is a novel pattern of paradoxically rapid tumor progression, which often leads to early death, mostly in the first 2 months of treatment with immune checkpoint inhibitors (ICIs). Currently, there is no validated biomarker to assess patients at risk of HPD. Aim The aim of this study was to systematically evaluate the predictive value of the neutrophil-to-lymphocyte ratio (NLR) in HPD and establish a reliable variable to support clinicians in defining personalized treatment strategies. Methods PubMed, Embase, Web of Science, Scopus, and Cochrane Library databases were searched for studies published before 31 December 2023. The Newcastle-Ottawa Scale (NOS) was used to evaluate the quality of eligible studies. The pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated using a random-effects or a fixed-effects model to evaluate the association between the NLR and the risk of HPD. Results A total of 17 studies with 2,964 patients were included for meta-analysis. The incidence of HPD across different types of tumors ranged from 6.3% to 35.6%. In the pooled analysis of the NLR and HPD, we identified that the NLR significantly associated with the risk of HPD (OR = 0.65; 95% CI: 0.46 to 0.91; p = 0.01) (I 2 = 52%, p = 0.007). Conclusion In the future, the NLR may serve as a remarkable biomarker for predicting the risk of HPD in clinical practice.
Collapse
Affiliation(s)
- Bo Pei
- Department of Oncology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, China
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jue Zhang
- Department of Radiology, The First People’s Hospital of Tianmen City, Tianmen, China
| | - Lin Lai
- Department of Oncology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, China
| | - Hui Chen
- Department of Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| |
Collapse
|
4
|
Nishimura K, Takahara K, Komura K, Ishida M, Hirosuna K, Maenosono R, Ajiro M, Sakamoto M, Iwatsuki K, Nakajima Y, Tsujino T, Taniguchi K, Tanaka T, Inamoto T, Hirose Y, Ono F, Kondo Y, Yoshimi A, Azuma H. Mechanistic insights into lethal hyper progressive disease induced by PD-L1 inhibitor in metastatic urothelial carcinoma. NPJ Precis Oncol 2024; 8:206. [PMID: 39289546 PMCID: PMC11408499 DOI: 10.1038/s41698-024-00707-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024] Open
Abstract
Hyper progressive disease (HPD) is a paradoxical phenomenon characterized by accelerated tumor growth following treatment with immune checkpoint inhibitors. However, the pathogenic causality and its predictor remain unknown. We herein report a fatal case of HPD in a 50-year-old man with metastatic bladder cancer. He had achieved a complete response (CR) through chemoradiation therapy followed by twelve cycles of chemotherapy, maintaining CR for 24 months. Three weeks after initiating maintenance use of a PD-L1 inhibitor, avelumab, a massive amount of metastases developed, leading to the patient's demise. Omics analysis, utilizing metastatic tissues obtained from an immediate autopsy, implied the contribution of M2 macrophages, TGF-β signaling, and interleukin-8 to HPD pathogenesis.
Collapse
Affiliation(s)
- Kazuki Nishimura
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
- Division of Cancer RNA Research, National Cancer Center Research Institute, Chuo-Ku, Tokyo, Japan
| | - Kiyoshi Takahara
- Department of Urology, Fujita-Health University School of Medicine, Toyoake City, Aichi, Japan
| | - Kazumasa Komura
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan.
- Division of Translational Research, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan.
| | - Mitsuaki Ishida
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Kensuke Hirosuna
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama City, Okayama, Japan
| | - Ryoichi Maenosono
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
- Division of Cancer RNA Research, National Cancer Center Research Institute, Chuo-Ku, Tokyo, Japan
| | - Masahiko Ajiro
- Division of Cancer RNA Research, National Cancer Center Research Institute, Chuo-Ku, Tokyo, Japan
| | - Moritoshi Sakamoto
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
- Division of Cancer RNA Research, National Cancer Center Research Institute, Chuo-Ku, Tokyo, Japan
| | - Kengo Iwatsuki
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Yuki Nakajima
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Takuya Tsujino
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Kohei Taniguchi
- Division of Translational Research, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Tomohito Tanaka
- Division of Translational Research, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Teruo Inamoto
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Yoshinobu Hirose
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Fumihito Ono
- Division of Translational Research, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Yoichi Kondo
- Department of Anatomy and Cell Biology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Akihide Yoshimi
- Division of Cancer RNA Research, National Cancer Center Research Institute, Chuo-Ku, Tokyo, Japan.
| | - Haruhito Azuma
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| |
Collapse
|
5
|
Zhang W, Chen X, Chen X, Li J, Wang H, Yan X, Zha H, Ma X, Zhao C, Su M, Hong L, Li P, Ling Y, Zhao W, Xia Y, Li B, Zheng T, Gu J. Fc-Fc interactions and immune inhibitory effects of IgG4: implications for anti-PD-1 immunotherapies. J Immunother Cancer 2024; 12:e009034. [PMID: 38925680 PMCID: PMC11203076 DOI: 10.1136/jitc-2024-009034] [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] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The majority of anti-programmed cell-death 1 (PD-1) monoclonal antibodies (mAbs) use S228P mutation IgG4 as the structural basis to avoid the activation of immune cells or complement. However, little attention has been paid to the Fc-Fc interactions between IgG4 and other IgG Fc fragments that could result in adverse effects. Fc-null IgG1 framework is a potential safer alternative to avoid the undesirable Fc-Fc interactions and Fc receptor binding derived effects observed with IgG4. This study provides a comprehensive evaluation of anti-PD-1 mAbs of these two frameworks. METHODS Trastuzumab and rituximab (both IgG1), wildtype IgG1 and IgG4 were immobilized on nitrocellulose membranes, coated to microplates and biosensor chips, and bound to tumor cells as targets for Fc-Fc interactions. Wildtype IgG1 and IgG4, anti-PD-1 mAb nivolumab (IgG4 S228P), penpulimab (Fc-null IgG1), and tislelizumab (Fc-null IgG4 S228P-R409K) were assessed for their binding reactions to the immobilized IgG proteins and quantitative kinetic data were obtained. To evaluate the effects of the two anti-PD-1 mAbs on immune responses mediated by trastuzumab and rituximab in the context of combination therapy, we employed classic immune models for antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and complement dependent cytotoxicity. Tumor-bearing mouse models, both wildtype and humanized, were used for in vivo investigation. Furthermore, we also examined the effects of IgG1 and IgG4 on diverse immune cell populations RESULTS: Experiments demonstrated that wildtype IgG4 and nivolumab bound to immobilized IgG through Fc-Fc interactions, diminishing antibody-dependent cell-mediated cytotoxicity and phagocytosis reactions. Quantitative analysis of kinetic parameters suggests that nivolumab and wildtype IgG4 exhibit comparable binding affinities to immobilized IgG1 in both non-denatured and denatured states. IgG4 exerted inhibitory effects on various immune cell types. Wildtype IgG4 and nivolumab both promoted tumor growth in wildtype mouse models. Conversely, wildtype IgG1, penpulimab, and tislelizumab did not show similar adverse effects. CONCLUSIONS Fc-null IgG1 represents a safer choice for anti-PD-1 immunotherapies by avoiding both the adverse Fc-Fc interactions and Fc-related immune inhibitory effects of IgG4. Fc-null IgG4 S228P-R409K and Fc-null IgG1 displayed similar structural properties and benefits. This study contributes to the understanding of immunotherapy resistance and the advancement of safer immune therapies for cancer.
Collapse
Affiliation(s)
- Weifeng Zhang
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Xueling Chen
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Xingxing Chen
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Jirui Li
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Hui Wang
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Xiaomiao Yan
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
- Jinxin Research Institute for Reproductive Medicine and Genetics, Xinan Hospital for Maternal and Child Health Care, Chengdu, China
| | - Han Zha
- The People's Hospital of Qijiang District Chongqing, Chongqing, China
| | - Xiaonan Ma
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Chanyuan Zhao
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Meng Su
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Liangli Hong
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Penghao Li
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
- Jinxin Research Institute for Reproductive Medicine and Genetics, Xinan Hospital for Maternal and Child Health Care, Chengdu, China
| | - Yanyu Ling
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Wenhui Zhao
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Yu Xia
- Akeso Biopharma Inc, Zhongshan, China
| | | | - Tianjing Zheng
- Chia Tai Tianqing Pharmaceutical Group Co., LTD, Nanjing, China
| | - Jiang Gu
- Guangdong Provincial International Collaborative Center of Molecular Medicine, Center of Collaboration and Creative, Molecular Diagnosis and Personalized Medical, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
- Jinxin Research Institute for Reproductive Medicine and Genetics, Xinan Hospital for Maternal and Child Health Care, Chengdu, China
| |
Collapse
|
6
|
Zhou Y, Ma B, Gao Q, Zhao L. The efficacy of subsequent therapy after failure of anti-PD-1 antibody in metastatic renal cell carcinoma. Transl Cancer Res 2024; 13:2238-2250. [PMID: 38881916 PMCID: PMC11170537 DOI: 10.21037/tcr-23-2390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/11/2024] [Indexed: 06/18/2024]
Abstract
Background The optional regimens of subsequent therapy after failure of anti-programmed cell death protein-1 (PD-1) antibody in metastatic renal cell carcinoma (mRCC) remain to be explored. There are reports of the efficacy of single-agent vascular endothelial growth factor receptor tyrosine kinase inhibitor (VEGFR-TKI) in patients with mRCC after failure of anti-PD-1 antibody therapy. However, it is not clear whether it is beneficial for patients to receive anti-PD-1 antibody as post-progression treatment. It has great significance to explore whether continuous application of anti-PD-1 antibody is beneficial for patients with mRCC whose diseases progressed to the state of pre-anti-PD-1 therapy. The purposes of this study are to explore the efficacy and safety of subsequent treatment on whether to continue using anti-PD-1 antibody therapy for patients who have progressive mRCC after prior treatment with anti-PD-1 antibody. Methods The clinical data of patients with mRCC from the Department of Immunotherapy in the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital from February 1, 2019 to December 31, 2021 were analyzed retrospectively. The primary endpoints were the objective response rate (ORR) and progression-free survival (PFS). The ORR and disease control rate (DCR) were estimated with Fisher's exact test. PFS and overall survival (OS) were assessed using the Kaplan-Meier method and log-rank tests. The associations between potential prognostic variables and PFS were evaluated with univariate and multivariate Cox regression analyses. A P value of less than or equal to 0.05 was deemed as statistically significant. Results A total of 35 patients were included in this study, during which 19 received VEGFR-TKI monotherapy and 16 received the VEGFR-TKI plus anti-PD-1 antibody therapy. Until the last follow-up on June 30, 2022, 19 patients experienced progressive disease (PD), five were in remission, and 11 kept stable disease (SD). After a median follow-up of 28.7 [95% confidence interval (CI): 17.0-35.6] months, the median PFS (mPFS) was 11.6 months for the VEGFR-TKI group and 9.1 months for the VEGFR-TKI plus anti-PD-1 antibody group [hazard ratio (HR) =0.81, 95% CI: 0.32-1.03, P=0.44]. Median OS (mOS) were 16.9 and 11.2 months respectively (HR =0.99, 95% CI: 0.44-2.27, P=0.90). The ORRs were 26.3% and 0% (P=0.049), and the DCRs were 47.4% and 43.8% (P=0.55) respectively. Treatment-related adverse events (TRAEs) occurred in 14 patients (73.7%) in the VEGFR-TKI group and 14 patients (87.5%) in the VEGFR-TKI plus anti-PD-1 antibody group (P=0.42); grade 3/4 TRAEs occurred in two patients (10.5%) and six patients (37.5%) respectively (P=0.11). Conclusions VEGFR-TKI monotherapy is an efficacious regimen for patients with mRCC whose diseases progressed on previous anti-PD-1 antibody therapy, and continuous anti-PD-1 therapy after failure of anti-PD-1 antibody could not provide additional clinical benefit but increased the incidence of TRAEs.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Baozhen Ma
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Quanli Gao
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Lingdi Zhao
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
7
|
Ren Z, Yang K, Zhu L, Yin D, Zhou Y. Regulatory T cells as crucial trigger and potential target for hyperprogressive disease subsequent to PD-1/PD-L1 blockade for cancer treatment. Int Immunopharmacol 2024; 132:111934. [PMID: 38574701 DOI: 10.1016/j.intimp.2024.111934] [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: 12/20/2023] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/06/2024]
Abstract
PD-1/PD-L1 blockade therapy has brought great success to cancer treatment. Nevertheless, limited beneficiary populations and even hyperprogressive disease (HPD) greatly constrain the application of PD-1/PD-L1 inhibitors in clinical treatment. HPD is a special pattern of disease progression with rapid tumor growth and even serious consequences of patient death, which requires urgent attention. Among the many predisposing causes of HPD, regulatory T cells (Tregs) are suspected because they are amplified in cases of HPD. Tregs express PD-1 thus PD-1/PD-L1 blockade therapy may have an impact on Tregs which leads to HPD. Tregs are a subset of CD4+ T cells expressing FoxP3 and play critical roles in suppressing immunity. Tregs migrate toward tumors in the presence of chemokines to suppress antitumor immune responses, causing cancer cells to grow and proliferate. Studies have shown that deleting Tregs could enhance the efficacy of PD-1/PD-L1 blockade therapy and reduce the occurrence of HPD. This suggests that immunotherapy combined with Treg depletion may be an effective means of avoiding HPD. In this review, we summarized the immunosuppressive-related functions of Tregs in antitumor therapy and focused on advances in therapy combining Tregs depletion with PD-1/PD-L1 blockade in clinical studies. Moreover, we provided an outlook on Treg-targeted HPD early warning for PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Zhe Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Kaiqing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Lin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Detao Yin
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
8
|
Song L, Gong Y, Wang E, Huang J, Li Y. Unraveling the tumor immune microenvironment of lung adenocarcinoma using single-cell RNA sequencing. Ther Adv Med Oncol 2024; 16:17588359231210274. [PMID: 38606165 PMCID: PMC11008351 DOI: 10.1177/17588359231210274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 10/09/2023] [Indexed: 04/13/2024] Open
Abstract
Tumor immune microenvironment (TIME) and its indications for lung cancer patient prognosis and therapeutic response have become new hotspots in cancer research in recent years. Tumor cells, immune cells, various regulatory factors, and their interactions in the TIME have been suggested to commonly influence lung cancer development and therapeutic outcome. The heterogeneity of TIME is composed of dynamic immune-related components, including various cancer cells, immune cells, cytokine/chemokine environments, cytotoxic activity, or immunosuppressive factors. The specific composition of cell subtypes may facilitate or hamper the response to immunotherapy and influence patient prognosis. Various markers have been found to stratify the patient prognosis or predict the therapeutic outcome. In this article, we systematically reviewed the recent advancement of TIME studies in lung adenocarcinoma (LUAD) using single-cell RNA sequencing (scRNA-seq) techniques, with specific focuses on the roles of TIME in LUAD development, TIME heterogeneity, indications of TIME in patient prognosis and therapeutic response during immunotherapy and drug resistance. The main findings in TIME heterogeneity and relevant markers or models for prognosis stratification and response prediction have been summarized. We hope that this review provides an overview of TIME status in LUAD and an inspiration for future development of strategies and biomarkers in LUAD treatment.
Collapse
Affiliation(s)
- Lele Song
- Department of Oncology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Yuan Gong
- Department of Gastroenterology, The Second Medical Center of the Chinese PLA General Hospital, Beijing, P.R. China
| | - Erpeng Wang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong province, P.R. China
| | - Jianchun Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University. No. 295, Xichang Road, Wuhua District, Kunming, Yunnan Province 650032, P.R. China
| | - Yuemin Li
- Department of Oncology, Chinese PLA General Hospital. No.8, Dongdajie, Fengtai District, Beijing 100071, P.R. China
| |
Collapse
|
9
|
Suzuki N, Matsuoka A, Horiuchi M, Sasaki A, Motomura Y. Rapid tumor progression complicated with liver abscess in a patient with gastric cancer receiving nivolumab therapy. Int Cancer Conf J 2024; 13:119-123. [PMID: 38524660 PMCID: PMC10957815 DOI: 10.1007/s13691-023-00647-1] [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: 09/27/2023] [Accepted: 12/13/2023] [Indexed: 03/26/2024] Open
Abstract
Immune checkpoint inhibitors have been approved for treating various cancer types. However, several studies reported rapid tumor progression, a condition known as hyperprogressive disease, after treatment with immune checkpoint inhibitors. We present the case of a 73-year-old man diagnosed with recurrent gastric cancer with liver and lymph node metastases detected in the presence of obstructive jaundice. Concomitant administration of nivolumab with cytotoxic chemotherapy as first-line chemotherapy effectively controlled the tumor. Nevertheless, once cytotoxic chemotherapy was discontinued and nivolumab monotherapy was initiated to treat liver abscess complications, the tumor rapidly progressed, ultimately leading to the patient's death. This is the first report on rapid tumor growth observed during subsequent treatment with nivolumab after initial antitumor effects were confirmed. This case report describes the possibility of rapid tumor growth in patients receiving immune checkpoint inhibitor therapy, including in cases where this therapy showed antitumor efficacy in the initial therapeutic evaluation. Therefore, patients receiving immune checkpoint inhibitor therapy need to be monitored.
Collapse
Affiliation(s)
- Natsumi Suzuki
- Department of Gastroenterology, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba 279-0001 Japan
- Department of General Internal Medicine, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba 279-0001 Japan
| | - Anna Matsuoka
- Department of General Internal Medicine, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba 279-0001 Japan
| | - Masao Horiuchi
- Department of Gastroenterology, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba 279-0001 Japan
| | - Akinori Sasaki
- Department of Gastroenterology, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba 279-0001 Japan
- Department of Oncology, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba 279-0001 Japan
| | - Yasuaki Motomura
- Department of Gastroenterology, Tokyo Bay Urayasu Ichikawa Medical Center, 3-4-32 Toudaijima, Urayasu, Chiba 279-0001 Japan
| |
Collapse
|
10
|
Wang ZQ, Wu ZX, Wang ZP, Bao JX, Wu HD, Xu DY, Li HF, Xu YY, Wu RX, Dai XX. Pan-cancer analysis of NUP155 and validation of its role in breast cancer cell proliferation, migration, and apoptosis. BMC Cancer 2024; 24:353. [PMID: 38504158 PMCID: PMC10953186 DOI: 10.1186/s12885-024-12039-6] [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: 10/19/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
NUP155 is reported to be correlated with tumor development. However, the role of NUP155 in tumor physiology and the tumor immune microenvironment (TIME) has not been previously examined. This study comprehensively investigated the expression, immunological function, and prognostic significance of NUP155 in different cancer types. Bioinformatics analysis revealed that NUP155 was upregulated in 26 types of cancer. Additionally, NUP155 upregulation was strongly correlated with advanced pathological or clinical stages and poor prognosis in several cancers. Furthermore, NUP155 was significantly and positively correlated with DNA methylation, tumor mutational burden, microsatellite instability, and stemness score in most cancers. Additionally, NUP155 was also found to be involved in TIME and closely associated with tumor infiltrating immune cells and immunoregulation-related genes. Functional enrichment analysis revealed a strong correlation between NUP155 and immunomodulatory pathways, especially antigen processing and presentation. The role of NUP155 in breast cancer has not been examined. This study, for the first time, demonstrated that NUP155 was upregulated in breast invasive carcinoma (BRCA) cells and revealed its oncogenic role in BRCA using molecular biology experiments. Thus, our study highlights the potential value of NUP155 as a biomarker in the assessment of prognostic prediction, tumor microenvironment and immunotherapeutic response in pan-cancer.
Collapse
Affiliation(s)
- Zi-Qiong Wang
- Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, 100 Minjiang Avenue, Quzhou, Zhejiang, 324000, Zhejiang, China
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhi-Xuan Wu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zong-Pan Wang
- Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, 100 Minjiang Avenue, Quzhou, Zhejiang, 324000, Zhejiang, China
| | - Jing-Xia Bao
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Hao-Dong Wu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Di-Yan Xu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Hong-Feng Li
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yi-Yin Xu
- Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, 100 Minjiang Avenue, Quzhou, Zhejiang, 324000, Zhejiang, China
| | - Rong-Xing Wu
- Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, 100 Minjiang Avenue, Quzhou, Zhejiang, 324000, Zhejiang, China.
| | - Xuan-Xuan Dai
- Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, 100 Minjiang Avenue, Quzhou, Zhejiang, 324000, Zhejiang, China.
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
11
|
Qin D, Zhang Y, Shu P, Lei Y, Li X, Wang Y. Targeting tumor-infiltrating tregs for improved antitumor responses. Front Immunol 2024; 15:1325946. [PMID: 38500876 PMCID: PMC10944859 DOI: 10.3389/fimmu.2024.1325946] [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: 10/22/2023] [Accepted: 02/16/2024] [Indexed: 03/20/2024] Open
Abstract
Immunotherapies have revolutionized the landscape of cancer treatment. Regulatory T cells (Tregs), as crucial components of the tumor immune environment, has great therapeutic potential. However, nonspecific inhibition of Tregs in therapies may not lead to enhanced antitumor responses, but could also trigger autoimmune reactions in patients, resulting in intolerable treatment side effects. Hence, the precision targeting and inhibition of tumor-infiltrating Tregs is of paramount importance. In this overview, we summarize the characteristics and subpopulations of Tregs within tumor microenvironment and their inhibitory mechanisms in antitumor responses. Furthermore, we discuss the current major strategies targeting regulatory T cells, weighing their advantages and limitations, and summarize representative clinical trials targeting Tregs in cancer treatment. We believe that developing therapies that specifically target and suppress tumor-infiltrating Tregs holds great promise for advancing immune-based therapies.
Collapse
Affiliation(s)
- Diyuan Qin
- Cancer Center, Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Cancer Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yugu Zhang
- Cancer Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Pei Shu
- Cancer Center, Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Cancer Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanna Lei
- Cancer Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoyu Li
- Cancer Center, Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Cancer Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongsheng Wang
- Cancer Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Wang X, Zhang L, Liao L, Li N, Tang T, Sun J, Zhou Z, Liu Y, Huang J, Wang Y, Chen Z, Zhang H, Xiao T, Tian Y, Zheng X, Yuan Y, Xiao L, Liu L, Guan J. SAA1 and metabolomic signatures predict hyperprogression with immunotherapy in pan cancers. Clin Transl Med 2024; 14:e1624. [PMID: 38468504 PMCID: PMC10928447 DOI: 10.1002/ctm2.1624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Affiliation(s)
- Xiaoqing Wang
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Longshan Zhang
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Liwei Liao
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Nan Li
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Tingxi Tang
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Jianda Sun
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Department of Radiation OncologyMeizhou People's HospitalMeizhouChina
| | - Zhenhua Zhou
- Department of Respiratory and Critical Care MedicineChronic Airways Diseases LaboratoryNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yang Liu
- Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangzhouChina
| | - Jihong Huang
- Department of Respiratory and Critical Care MedicineChronic Airways Diseases LaboratoryNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yingqiao Wang
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Zekai Chen
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Hanbin Zhang
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Ting Xiao
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yunming Tian
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Xiuting Zheng
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yi Yuan
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Linlin Xiao
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Laiyu Liu
- Department of Respiratory and Critical Care MedicineChronic Airways Diseases LaboratoryNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Jian Guan
- Department of Radiation OncologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Guangdong Province Key Laboratory of Molecular Tumor PathologyGuangzhouChina
| |
Collapse
|
13
|
Alkader MS, Altaha RZ, Jabali EH, Attieh OA, Matalqa AW. Is there an association between lymph node size and hyperprogression in immunotherapy-treated patients? ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2024; 62:33-43. [PMID: 37882575 DOI: 10.2478/rjim-2023-0025] [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: 07/22/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND Hyperprogressive disease (HPD) can be described as an accelerated increase in the growth rate of tumors combined with rapid clinical deterioration observed in a subset of cancer patients undergoing immunotherapy, specifically with immune checkpoint inhibitors (ICIs). The reported incidence of HPD ranges from 5.9% to 43.1% in patients receiving ICIs. In this context, identifying reliable predictive risk factors for HPD is crucial as it may allow for earlier intervention and ultimately improve patient outcomes. METHODS This study retrospectively analyzed ten metastatic renal cell carcinoma (mRCC) patients. The identification of HPD was based on the diagnostic criteria proposed by Ferrara R et al. This study aimed to investigate whether there is an association between LN size and HPD using a cutoff value of 3 cm for LN size. Given the limited sample size, Fisher's exact test was used to test this association. We conducted a Kaplan-Meier (KM) analysis to estimate the median overall survival (OS) of patients with HPD and compared it to those without HPD. RESULTS Three patients (30%) developed HPD, while seven (70%) did not. Fisher's exact test revealed a statistically significant association between the HPD and LN size ≥ 3 cm (p=0.008). In the HPD group, the median OS was significantly shorter, with a median OS of 3 months, whereas in the non-HPD group, the median OS was not reached (P =0.001). CONCLUSION The present study found a significant association between LN size ≥ 3 cm in the pretreatment period and HPD development.
Collapse
Affiliation(s)
- Mohammad S Alkader
- Department of Clinical Oncology, Military Cancer Center, Royal Medical Services, Amman, Jordan
| | - Rashed Z Altaha
- Department of Internal Medicine, Military Cancer Center, Royal Medical Services, Amman, Jordan
| | - Eslam H Jabali
- Department of nuclear medicine, Royal Medical Services, Amman, Jordan
| | - Ola A Attieh
- Department of nuclear medicine, Royal Medical Services, Amman, Jordan
| | - Ala' W Matalqa
- Department of Internal Medicine, Military Cancer Center, Royal Medical Services, Amman, Jordan
| |
Collapse
|
14
|
Liu Q, Zhang C, Chen X, Han Z. Modern cancer therapy: cryoablation meets immune checkpoint blockade. Front Oncol 2024; 14:1323070. [PMID: 38384806 PMCID: PMC10881233 DOI: 10.3389/fonc.2024.1323070] [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: 10/17/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
Cryoablation, as a minimally invasive technology for the treatment of tumors, destroys target tumors with lethal low temperatures. It simultaneously releases a large number of tumor-specific antigens, pro-inflammatory cytokines, and nucleoproteins, known as "danger signals", activating the body's innate and adaptive immune responses. However, tumor cells can promote the inactivation of immune effector cells by reprogramming immune checkpoints, leading to the insufficiency of these antigens to induce an immune response capable of eradicating the tumor. Immune checkpoint blockers rejuvenate exhausted T cells by blocking immune checkpoints that induce programmed death of T cells, and are therefore considered a promising therapeutic strategy to enhance the immune effects of cryoablation. In this review, we provide a detailed explanation of the immunological mechanisms of cryoablation and articulate the theoretical basis and research progress of the treatment of cancer with cryoablation combined with immune checkpoint blockers. Preliminary data indicates that this combined treatment strategy exhibits good synergy and has been proven to be safe and effective.
Collapse
Affiliation(s)
- Qi Liu
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Chunyang Zhang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xuxin Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
15
|
Kato M, Uchida J. Recent advances in immune checkpoint inhibitors in the treatment of urothelial carcinoma: A review. Int J Urol 2023; 30:1068-1077. [PMID: 37602512 DOI: 10.1111/iju.15278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023]
Abstract
Treatment options for urothelial carcinoma were limited until the emergence of immune checkpoint inhibitors, and even now, the prognosis of metastatic disease is poor compared with the other two major genitourinary cancers, renal cell carcinoma and prostate cancer. Despite the increasing use of immune checkpoint inhibitors in the sequential treatment of urothelial carcinoma, conflicting results from similar randomized clinical trials call into question the efficacy of this treatment. In addition, physicians must be aware of the clinical characteristics of immune checkpoint inhibitors, including immune-related adverse events, pseudo- and hyperprogression. This review summarizes the conflicting results of recent clinical trials and provides insights into the role of immune checkpoint inhibitors in the treatment of urothelial carcinoma.
Collapse
Affiliation(s)
- Minoru Kato
- Department of Urology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Junji Uchida
- Department of Urology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
16
|
Zhou R, Tong F, Zhang Y, Zhang R, Bin Y, Zhang S, Yang N, Dong X. Genomic alterations associated with pseudoprogression and hyperprogressive disease during anti-PD1 treatment for advanced non-small-cell lung cancer. Front Oncol 2023; 13:1231094. [PMID: 38023206 PMCID: PMC10667039 DOI: 10.3389/fonc.2023.1231094] [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: 05/30/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction This study aimed to elucidate the relationship between dynamic genomic mutation alteration and pseudoprogression (PsPD)/hyperprogressive disease (HPD) in immunotherapy-treated advanced non-small-cell lung cancer (NSCLC), to provide clinical evidence for identifying and distinguishing between PsPD and HPD. Method Patients with advanced NSCLC who were treated with anti-PD1 were enrolled. Whole blood was collected at baseline and post image progression. Serum was separated and sequenced using 425-panel next-generation sequencing analysis (NGS). Results NGS revealed that not only single gene mutations were associated with PsPD/HPD before treatment, dynamic monitoring of the whole-blood genome mutation spectrum also varied greatly. Mutational burden, allele frequency%, and relative circulating tumor DNA abundance indicated that the fold change after image progression was much higher in the HPD group. Discussion The gene mutation profiles of PsPD and HPD not only differed before treatment, but higher genome mutation spectrum post image progression indicated true disease progression in patients with HPD. This suggests that dynamic whole-genome mutation profile monitoring as NGS can distinguish PsPD from HPD more effectively than single gene detection, providing a novel method for guiding clinical immune treatment.
Collapse
Affiliation(s)
- Rui Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Tong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongchang Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruigang Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yawen Bin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nong Yang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Yan D. Hope and Challenges: Immunotherapy in EGFR-Mutant NSCLC Patients. Biomedicines 2023; 11:2916. [PMID: 38001917 PMCID: PMC10669068 DOI: 10.3390/biomedicines11112916] [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: 09/05/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
EGFR tyrosine kinase inhibitors (TKIs) are the preferred initial treatment for non-small cell lung cancer (NSCLC) patients harboring sensitive EGFR mutations. Sadly, remission is transient, and no approved effective treatment options are available for EGFR-TKI-advanced EGFR-mutant NSCLCs. Although immunotherapy with immune checkpoint inhibitors (ICIs) induces sustained cancer remission in a subset of NSCLCs, ICI therapy exhibits limited activity in most EGFR-mutant NSCLCs. Mechanistically, the strong oncogenic EGFR signaling in EGFR-mutant NSCLCs contributes to a non-inflamed tumor immune microenvironment (TIME), characterized by a limited number of CD8+ T cell infiltration, a high number of regulatory CD4+ T cells, and an increased number of inactivated infiltrated T cells. Additionally, EGFR-mutant NSCLC patients are generally non-smokers with low levels of PD-L1 expression and tumor mutation burden. Promisingly, a small population of EGFR-mutant NSCLCs still durably respond to ICI therapy. The hope of ICI therapy from pre-clinical studies and clinical trials is reviewed in EGFR-mutant NSCLCs. The challenges of application ICI therapy in EGFR-mutant NSCLCs are also reviewed.
Collapse
Affiliation(s)
- Dan Yan
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA;
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
18
|
Xu S, Wang W, Meng T, Wang F, Wang G, Huang F, Wang G, Yu X, Wu R, Hou L, Ye Z, Zhang X, Zhao H, Shen Y. Construction and validation of a immune-related prognostic gene DHRS1 in hepatocellular carcinoma based on bioinformatic analysis. Medicine (Baltimore) 2023; 102:e35268. [PMID: 37861541 PMCID: PMC10589603 DOI: 10.1097/md.0000000000035268] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023] Open
Abstract
A member of the short-chain dehydrogenase/reductase superfamily (DHRS1, SDR19C1) is a member of the short-chain dehydrogenase/reductase superfamily and a potential predictor of hepatocellular carcinoma (HCC). However, the role of DHRS1 in HCC immunity remains unclear. We systematically analyzed the association between DHRS1 and HCC immunity with transcriptional and clinical data from the Tumor Immune Estimation Resource, an integrated repository portal for tumor immune system interactions, and cBioPortal databases. Six DHRS1-associated immunomodulators strongly correlated with survival and were uncovered by exploiting univariate and multivariate Cox analyses. We created a risk score for each patient by adding the points from each immunomodulator and then classified them into high and low risk categories. Survival analysis were used to compare the overall survival between the 2 groups, and the receiver operating characteristic curve was applied to assess the accuracy of the risk score. Data from our center were adopted as the external validation set, the risk score was calculated using the risk coefficient of the 6 genes in the training cohort, and survival analysis were executed to verify the experimental group results. A nomogram was ultimately constructed with the R package. Our data revealed a correlation between the levels of immune cell infiltration and either the DHRS1 gene copy numbers or mRNA levels in HCC. Second, we generated a signature based on the 6 DHRS1-related immunomodulators (KDR, TNFRSF4, CD276, TNFSF4, SLAMF6, and SIGLEC9). We postulate that the generated risk scores would serve as an independent indicator of HCC prognosis, with an area under the receiver operating characteristic curve for the risk score of 0.743. We further established external validation sets to reconfirm the predictive validity of the risk score. Finally, a prognostic nomogram and calibration curve were created. The DHRS1 gene may exert an impact on HCC immunity. We posit that the nominated immune signature based on DHRS1-associated immunomodulators could constitute a promising prognostic biomarker in HCC.
Collapse
Affiliation(s)
- Sa Xu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Wei Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Organ Transplant Center of The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tao Meng
- Department of General Surgery, Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fuyan Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Guoxing Wang
- Anhui BioX-Vision Biological Technology Co., Ltd, Hefei, China
| | - Fan Huang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Organ Transplant Center of The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guobin Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Organ Transplant Center of The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaojun Yu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Organ Transplant Center of The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ruolin Wu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Organ Transplant Center of The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liujin Hou
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Organ Transplant Center of The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhenghui Ye
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Organ Transplant Center of The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinghua Zhang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Organ Transplant Center of The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongchuan Zhao
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Organ Transplant Center of The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
19
|
Pan Y, Ma Y, Dai G. The Prognostic Value of the Prognostic Nutritional Index in Patients with Advanced or Metastatic Gastric Cancer Treated with Immunotherapy. Nutrients 2023; 15:4290. [PMID: 37836573 PMCID: PMC10574242 DOI: 10.3390/nu15194290] [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: 09/13/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
In recent years, the therapeutic effect of monoclonal antibodies against programmed cell death protein-1 (PD-1) in patients with locally advanced or metastatic gastric or gastroesophageal junction (G/GEJ) cancer has been confirmed in many studies. The exploration and discovery of new biomarker combinations based on tumor characteristics and tumor microenvironment help screen superior patients and realize precise immunotherapy. As an evaluation index of immunonutritional status, the prognostic nutritional index (PNI) is low cost, simple and easy to obtain, and effective in determining the prognosis of tumor patients. We selected 268 consecutive AGC patients who were treated with ICI therapy from December 2014 to May 2021. We measured their pretreatment of the PNI levels and performed univariate and multivariate Cox regression analyses of progression-free survival (PFS) or overall survival (OS) after ICI therapy. The low pretreatment PNI level of AGC patients was significantly correlated with shorter PFS (p < 0.001) and OS (p < 0.001) after ICI treatment. In univariate and multivariate analyses of the associations between PNI and OS or PFS, PNI is an independent prognostic factor for PFS (HR = 1.511; 95%CI 1.154-1.977; p = 0.003) and OS (HR = 1.431; 95%CI 1.049-1.951; p = 0.024), respectively. Notably, decreased PNI during treatment with ICIs was associated with early relapse and death. Pretreatment with PNI might help to identify AGC patients who will obtain a survival benefit from ICI therapy.
Collapse
Affiliation(s)
- Yuting Pan
- Chinese PLA Medical School, Beijing 100853, China; (Y.P.); (Y.M.)
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Yue Ma
- Chinese PLA Medical School, Beijing 100853, China; (Y.P.); (Y.M.)
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Guanghai Dai
- Chinese PLA Medical School, Beijing 100853, China; (Y.P.); (Y.M.)
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| |
Collapse
|
20
|
Long Y, Yang W, Bai Y, Tao H, Zhang F, Wang L, Yang B, Huang D, Han X, Hu Y. Prediction model for hyperprogressive disease in patients with advanced solid tumors received immune-checkpoint inhibitors: a pan-cancer study. Cancer Cell Int 2023; 23:224. [PMID: 37777758 PMCID: PMC10543870 DOI: 10.1186/s12935-023-03070-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/17/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Hyper progressive disease (HPD) describes the phenomenon that patients can't benefit from immunotherapy but cause rapid tumor progression. HPD is a particular phenomenon in immunotherapy but lacks prediction methods. Our study aims to screen the factors that may forecast HPD and provide a predictive model for risky stratifying. METHODS We retrospectively reviewed advanced-stage tumor patients who received immune checkpoint inhibitors (ICI) in the General PLA Hospital. Subsequently, we calculated the tumor growth kinetics ratio (TGKr) and identified typical HPD patients. Differences analysis of clinical characteristics was performed, and a predictive binary classification model was constructed. RESULTS 867 patients with complete image information were screened from more than 3000 patients who received ICI between January 2015 and January 2020. Among them, 36 patients were identified as HPD for TGKr > 2. After the propensity score matched, confounding factors were limited. Survival analysis revealed that the clinical outcome of HPD patients was significantly worse than non-HPD patients. Besides, we found that Body Mass Index (BMI), anemia, lymph node metastasis in non-draining areas, pancreatic metastasis, and whether combined with anti-angiogenesis or chemotherapy therapy were closely connected with the HPD incidence. Based on these risk factors, we constructed a visualised predicted nomogram model, and the Area Under Curve (AUC) is 0.850 in the train dataset, whereas 0.812 in the test dataset. CONCLUSION We carried out a retrospective study for HPD based on real-world patients and constructed a clinically feasible and practical model for predicting HPD incidence, which could help oncologists to stratify risky patients and select treatment strategies.
Collapse
Affiliation(s)
- Yaping Long
- School of Medicine, Nankai University, 94 Weijin Road, Nankai, Tianjin, China
| | - Wenyu Yang
- School of Medicine, Nankai University, 94 Weijin Road, Nankai, Tianjin, China
| | - Yibing Bai
- PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
- Department of Medical Oncology, Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, 8 Dongdajie Road, Fengtai District, Beijing, 100071, China
| | - Haitao Tao
- Department of Medical Oncology, Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, 8 Dongdajie Road, Fengtai District, Beijing, 100071, China
| | - Fan Zhang
- Department of Medical Oncology, Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, 8 Dongdajie Road, Fengtai District, Beijing, 100071, China
| | - Lijie Wang
- Department of Medical Oncology, Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, 8 Dongdajie Road, Fengtai District, Beijing, 100071, China
| | - Bo Yang
- Department of Medical Oncology, Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, 8 Dongdajie Road, Fengtai District, Beijing, 100071, China
| | - Di Huang
- Department of Medical Oncology, Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, 8 Dongdajie Road, Fengtai District, Beijing, 100071, China
| | - Xiao Han
- Department of Medical Oncology, Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, 8 Dongdajie Road, Fengtai District, Beijing, 100071, China.
| | - Yi Hu
- School of Medicine, Nankai University, 94 Weijin Road, Nankai, Tianjin, China.
- PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China.
- Department of Medical Oncology, Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, 8 Dongdajie Road, Fengtai District, Beijing, 100071, China.
| |
Collapse
|
21
|
Zhang Y, Han X, Wang K, Liu D, Ding X, Hu Z, Wang J. Co-Delivery Nanomicelles for Potentiating TNBC Immunotherapy by Synergetically Reshaping CAFs-Mediated Tumor Stroma and Reprogramming Immunosuppressive Microenvironment. Int J Nanomedicine 2023; 18:4329-4346. [PMID: 37545872 PMCID: PMC10403052 DOI: 10.2147/ijn.s418100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/17/2023] [Indexed: 08/08/2023] Open
Abstract
Purpose Immune checkpoint inhibitors (ICI) have received the most attention for triple negative breast cancer (TNBC), while the response rate to ICI remains limited due to insufficient T cell infiltration. It is therefore essential that alternative strategies are developed to improve the therapeutic outcomes of ICI in non-responsive TNBC cases. The efficacy of pH-responsive nanomicelles (P/A/B@NM) co-loaded with paclitaxel (PTX), CXCR4 antagonist AMD3100, and PD-1/PD-L1 inhibitor BMS-1 activating the T cell-mediated antitumor immune response were evaluated using a 4T1 antiPD-1-resistance breast tumor model. Methods In vitro, pH-responsive antitumor effect of P/A/B@NM was investigated by assessing cell viability, migration and invasion. In vivo, the distribution of P/A/B@NM was visualized in 4T1 orthotopic TNBC model using an IVIS spectrum imaging instrument. The efficacy of the co-delivery nanocarriers was evaluated by monitoring mouse survival, tumor growth and metastasis, cancer-associated fibroblasts (CAFs)-mediated tumor stroma and immunosuppressive microenvironment components, and the recruitment and infiltration of CD8+ T cells. Results The prepared P/A/B@NM in acid microenvironment demonstrates remarkable cytotoxicity against MDA-MB-231 cells, with an IC50 of 105 μg/mL. Additionally, it exhibits substantial inhibition of tumor cell migration and invasion. The P/A/B@NM based on co-delivery nanocarriers efficiently accumulate at the tumor site and release the drugs in a pH-responsive controlled manner. The nanomedicine-PTX, AMD3100, and BMS-1 formulation significantly inhibits tumor growth and lung/liver metastasis by inducing antitumor immune responses via CXCL12/CXCR4 axis blockade, and immunogenic cell death to reprogramme both tumor stroma and immunosuppressive microenvironment. As a result, CD8+ T cell infiltration is triggered into the tumor site, boosting the efficacy of ICI therapy synergistically. Conclusion These results demonstrate that combination therapy using P/A/B@NM reshapes CAFs-mediated tumor stroma and immunosuppressive microenvironment, which can enhance the infiltration of CD8+ T cells, thereby reactivating anti-tumor immunity for non-responsive TNBC cases.
Collapse
Affiliation(s)
- Yue Zhang
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Xue Han
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Ke Wang
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Da Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Xiaoyun Ding
- Oncology Hospital, General Hospital of Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Zhiqiang Hu
- Oncology Hospital, General Hospital of Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Jing Wang
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
- Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Yinchuan, 750004, People’s Republic of China
| |
Collapse
|
22
|
Yu D, Yang P, Lu X, Huang S, Liu L, Fan X. Single-cell RNA sequencing reveals enhanced antitumor immunity after combined application of PD-1 inhibitor and Shenmai injection in non-small cell lung cancer. Cell Commun Signal 2023; 21:169. [PMID: 37430270 DOI: 10.1186/s12964-023-01184-3] [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: 03/17/2023] [Accepted: 06/04/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have altered the clinical management of non-small cell lung cancer (NSCLC). However, the low response rate, severe immune-related adverse events (irAEs), and hyperprogressive disease following ICIs monotherapy require attention. Combination therapy may overcome these limitations and traditional Chinese medicine with immunomodulatory effects provides a promising approach. Shenmai injection (SMI) is a clinically effective adjuvant treatment for cancer with chemotherapy and radiotherapy. Therefore, the combined effects and mechanisms of SMI and programmed death-1 (PD-1) inhibitor against NSCLC was focused on this study. METHODS A Lewis lung carcinoma mouse model and a lung squamous cell carcinoma humanized mouse model were used to investigate the combined efficacy and safety of SMI and PD-1 inhibitor. The synergistic mechanisms of the combination therapy against NSCLC were explored using single-cell RNA sequencing. Validation experiments were performed using immunofluorescence analysis, in vitro experiment, and bulk transcriptomic datasets. RESULTS In both models, combination therapy alleviated tumor growth and prolonged survival without increasing irAEs. The GZMAhigh and XCL1high natural killer (NK) cell subclusters with cytotoxic and chemokine signatures increased in the combination therapy, while malignant cells from combination therapy were mainly in the apoptotic state, suggesting that mediating tumor cell apoptosis through NK cells is the main synergistic mechanisms of combination therapy. In vitro experiment confirmed that combination therapy increased secretion of Granzyme A by NK cells. Moreover, we discovered that PD-1 inhibitor and SMI combination blocked inhibitory receptors on NK and T cells and restores their antitumoral activity in NSCLC better than PD-1 inhibitor monotherapy, and immune and stromal cells exhibited a decrease of angiogenic features and attenuated cancer metabolism reprogramming in microenvironment of combination therapy. CONCLUSIONS This study demonstrated that SMI reprograms tumor immune microenvironment mainly by inducing NK cells infiltration and synergizes with PD-1 inhibitor against NSCLC, suggested that targeting NK cells may be an important strategy for combining with ICIs. Video Abstract.
Collapse
Affiliation(s)
- Dingyi Yu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Penghui Yang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Shaoze Huang
- Zhejiang Engineering Research Center for Advanced Manufacturing of Traditional Chinese Medicine, Huzhou, China
| | - Li Liu
- Zhejiang Engineering Research Center for Advanced Manufacturing of Traditional Chinese Medicine, Huzhou, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
| |
Collapse
|
23
|
Yin L, Wan Z, Sun P, Shuai P, Liu Y. Time to abandon CAR-T monotherapy for solid tumors. Biochim Biophys Acta Rev Cancer 2023; 1878:188930. [PMID: 37286147 DOI: 10.1016/j.bbcan.2023.188930] [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: 04/21/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023]
Abstract
In recent decades, chimeric antigen receptor T (CAR-T) cell therapy has achieved dramatic success in patients with hematological malignancies. However, CAR-T cell therapy failed to effectively treat solid tumors as a monotherapy. By summarizing the challenges of CAR-T cell monotherapy for solid tumors and analyzing the underlying mechanisms of combinatorial strategies to counteract these hurdles, we found that complementary therapeutics are needed to improve the scant and transient responses of CAR-T cell monotherapy in solid tumors. Further data, especially data from multicenter clinical trials regarding efficacy, toxicity, and predictive biomarkers are required before the CAR-T combination therapy can be translated into clinical settings.
Collapse
Affiliation(s)
- Limei Yin
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Zhengwei Wan
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Ping Sun
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Ping Shuai
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.
| | - Yuping Liu
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.
| |
Collapse
|
24
|
Gong C, Zhang W, Sun Y, Shou J, Jiang Z, Liu T, Wang S, Liu J, Sun Y, Zhou A. Exploration of the immunogenetic landscape of hyperprogressive disease after combined immunotherapy in cancer patients. iScience 2023; 26:106720. [PMID: 37255657 PMCID: PMC10225883 DOI: 10.1016/j.isci.2023.106720] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/31/2023] [Accepted: 04/19/2023] [Indexed: 06/01/2023] Open
Abstract
The immune-genetic changes that occur in cancer patients experiencing hyperprogressive disease (HPD) during combined immunotherapy are unclear. In this study, HPD patients with pre- and post-HPD samples and non-HPD patients with solid tumors were molecularly characterized by genetic and tumor immune microenvironment (TiME) analyses of paired samples by whole-exome sequencing, RNA sequencing, and multiplex immunofluorescence. The genetic analysis of paired samples showed that almost all the tumor driver gene mutations were preserved between pre- and post-HPD tumors. HPD patients had higher frequencies of mutations in TP53 and CNN2, and a significantly higher mutant-allele tumor heterogeneity than non-HPD patients. Tumor IL-6 mRNA was upregulated in post-HPD samples vs. pre-HPD, accompanied by a potential immune suppressive TiME with an elevated M2/M1 ratio. Salvage treatment with irinotecan plus bevacizumab was effective in one HPD patient, who experienced prolonged survival. These genetic features and TiME characteristics might help identify the features of HPD after immunotherapy.
Collapse
Affiliation(s)
- Caifeng Gong
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wen Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yongkun Sun
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jianzhong Shou
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhichao Jiang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Tianyi Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shengzhou Wang
- GenomiCare Biotechnology Co. Ltd, Shanghai 201203, China
| | - Jun Liu
- GenomiCare Biotechnology Co. Ltd, Shanghai 201203, China
| | - Ying Sun
- GenomiCare Biotechnology Co. Ltd, Shanghai 201203, China
| | - Aiping Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
25
|
Zheng J, Miao F, Wang Z, Ma Y, Lin Z, Chen Y, Kong X, Wang Y, Zhuang A, Wu T, Li W. Identification of MDM2 as a prognostic and immunotherapeutic biomarker in a comprehensive pan-cancer analysis: A promising target for breast cancer, bladder cancer and ovarian cancer immunotherapy. Life Sci 2023:121832. [PMID: 37276911 DOI: 10.1016/j.lfs.2023.121832] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND The murine double minute 2 (MDM2) gene is a crucial factor in the development and progression of various cancer types. Multiple rigorous scientific studies have consistently shown its involvement in tumorigenesis and cancer progression in a wide range of cancer types. However, a comprehensive analysis of the role of MDM2 in human cancer has yet to be conducted. METHODS We used various databases, including TIMER2.0, TCGA, GTEx and STRING, to analyze MDM2 expression and its correlation with clinical outcomes, interacting genes and immune cell infiltration. We also investigated the association of MDM2 with immune checkpoints and performed gene enrichment analysis using DAVID tools. RESULTS The pan-cancer MDM2 analysis found that MDM2 expression and mutation status were observably different in 25 types of cancer tissue compared with healthy tissues, and prognosis analysis showed that there was a significant correlation between MDM2 expression and patient prognosis. Furthermore, correlation analysis showed that MDM2 expression was correlated with tumor mutational burden, microsatellite instability and drug sensitivity in certain cancer types. We found that there was an association between MDM2 expression and immune cell infiltration across cancer types, and MDM2 inhibitors might enhance the effect of immunotherapy on breast cancer, bladder cancer and ovarian cancer. CONCLUSIONS The first systematic pan-cancer analysis of MDM2 was conducted, and it demonstrated that MDM2 was a reliable prognostic biomarker and was closely related to cancer immunity, providing a potential immunotherapeutic target for breast cancer, bladder cancer and ovarian cancer.
Collapse
Affiliation(s)
- Jialiang Zheng
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Fenglin Miao
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhao Wang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yuan Ma
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhenhang Lin
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yaqin Chen
- Nursing Department of Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Xu Kong
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yue Wang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Aobo Zhuang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Ting Wu
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Wengang Li
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
26
|
Wang J, Hong J, Yang F, Liu F, Wang X, Shen Z, Wu D. A deficient MIF-CD74 signaling pathway may play an important role in immunotherapy-induced hyper-progressive disease. Cell Biol Toxicol 2023; 39:1169-1180. [PMID: 34797429 DOI: 10.1007/s10565-021-09672-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/13/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND With the advent of immune checkpoint inhibitors (ICIs) therapies, a major breakthrough has been made in cancer treatment. However, instead of good results, some patients experienced a deterioration of their disease. This unexpected result is termed as hyper-progressive disease (HPD). The biology of HPD is currently not fully understood. METHODS Isolation of CD3+ cells from peripheral blood mononuclear cells (PBMC) in healthy control, tumor patients receiving immunotherapy with or without immunotherapy-induced HPD, then conducted single-cell RNA sequencing (scRNA-seq). RESULTS By analyzing scRNA-seq data, we identified 15 cell clusters. We observed developed-exhausted CD4+ T cells and regulatory T cells (Tregs) increasingly enriched in HPD group. Meanwhile, some effector T cells were decreased in HPD. The imbalance potentially contributes to the occurrence of HPD and poor clinical prognosis. In addition, we analyzed ligand-receptor interactions between subsets. The ligand-receptor interaction "CD74-MIF" was absent in HPD. However, in vitro experiment, we found that CD74 regulated effector function of effector CD8+ T cells. Overall, the article provides a primary study of immune profile in HPD.
Collapse
Affiliation(s)
- Jiahui Wang
- Department of Oncology, Affiliated Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jinsheng Hong
- Department of Radiotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Feiyu Yang
- Emergency Department of Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Fangming Liu
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiangdong Wang
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China.
| | - Zan Shen
- Department of Oncology, Affiliated Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Duojiao Wu
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
27
|
Laface C, Maselli FM, Santoro AN, Iaia ML, Ambrogio F, Laterza M, Guarini C, De Santis P, Perrone M, Fedele P. The Resistance to EGFR-TKIs in Non-Small Cell Lung Cancer: From Molecular Mechanisms to Clinical Application of New Therapeutic Strategies. Pharmaceutics 2023; 15:1604. [PMID: 37376053 DOI: 10.3390/pharmaceutics15061604] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/13/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Almost 17% of Western patients affected by non-small cell lung cancer (NSCLC) have an activating epidermal growth factor receptor (EGFR) gene mutation. Del19 and L858R are the most-common ones; they are positive predictive factors for EGFR tyrosine kinase inhibitors (TKIs). Currently, osimertinib, a third-generation TKI, is the standard first-line therapy for advanced NSCLC patients with common EGFR mutations. This drug is also administered as a second-line treatment for those patients with the T790M EGFR mutation and previously treated with first- (erlotinib, gefitinib) or second- (afatinib) generation TKIs. However, despite the high clinical efficacy, the prognosis remains severe due to intrinsic or acquired resistance to EGRF-TKIs. Various mechanisms of resistance have been reported including the activation of other signalling pathways, the development of secondary mutations, the alteration of the downstream pathways, and phenotypic transformation. However, further data are needed to achieve the goal of overcoming resistance to EGFR-TKIs, hence the necessity of discovering novel genetic targets and developing new-generation drugs. This review aimed to deepen the knowledge of intrinsic and acquired molecular mechanisms of resistance to EGFR-TKIs and the development of new therapeutic strategies to overcome TKIs' resistance.
Collapse
Affiliation(s)
- Carmelo Laface
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | | | | | - Maria Laura Iaia
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Francesca Ambrogio
- Section of Dermatology, Department of Biomedical Science and Human Oncology, University of Bari, 70124 Bari, Italy
| | - Marigia Laterza
- Division of Cardiac Surgery, University of Bari, 70124 Bari, Italy
| | - Chiara Guarini
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Pierluigi De Santis
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Martina Perrone
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Palma Fedele
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| |
Collapse
|
28
|
Li Y, Chen T, Nie TY, Han J, He Y, Tang X, Zhang L. Hyperprogressive disease in non-small cell lung cancer after PD-1/PD-L1 inhibitors immunotherapy: underlying killer. Front Immunol 2023; 14:1200875. [PMID: 37283759 PMCID: PMC10239849 DOI: 10.3389/fimmu.2023.1200875] [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: 04/05/2023] [Accepted: 04/24/2023] [Indexed: 06/08/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) target the negative regulatory pathway of T cells and effectively reactive the anti-tumor immune function of T cells by blocking the key pathway of the immune escape mechanism of the tumor-PD-1/PD-L1, and fundamentally changing the prospect of immunotherapy for non-small cell lung cancer patients. However, such promising immunotherapy is overshadowed by Hyperprogressive Disease, a response pattern associated with unwanted accelerated tumor growth and characterized by poor prognosis in a fraction of treated patients. This review comprehensively provides an overview of Hyperprogressive Disease in immune checkpoint inhibitor-based immunotherapy for non-small cell lung cancer including its definition, biomarkers, mechanisms, and treatment. A better understanding of the black side of immune checkpoint inhibitors therapy will provide a more profound insight into the pros and cons of immunotherapy.
Collapse
Affiliation(s)
- Yanping Li
- Department of Respiratory Medicine, The Third People’s Hospital of Honghe Prefecture, Gejiu, China
| | - Tianhong Chen
- Department of Thoracic Surgery , The Third People’s Hospital of Honghe Prefecture, Gejiu, China
| | - Tian Yi Nie
- Department of Respiratory Medicine, The Third People’s Hospital of Honghe Prefecture, Gejiu, China
| | - Juyuan Han
- Department of Respiratory Medicine, The Third People’s Hospital of Honghe Prefecture, Gejiu, China
| | - Yunyan He
- Department of Thoracic Surgery, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xingxing Tang
- Department of Thoracic Surgery , The Third People’s Hospital of Honghe Prefecture, Gejiu, China
| | - Li Zhang
- Department of Oncology, Gejiu City People’s Hospital, Diannan Central Hospital of Honghe Prefecture, The Fifth Affiliated Hospital of Kunming Medical University, Gejiu, China
| |
Collapse
|
29
|
Wang Y, Huang X, Fan H, Xu Y, Qi Z, Zhang Y, Huang Y. Identification of fatty acid-related subtypes, the establishment of a prognostic signature, and immune infiltration characteristics in lung adenocarcinoma. Aging (Albany NY) 2023; 15:204725. [PMID: 37199651 DOI: 10.18632/aging.204725] [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: 02/15/2023] [Accepted: 05/03/2023] [Indexed: 05/19/2023]
Abstract
Abnormal fatty acid (FA) metabolism can change the inflammatory microenvironment and promote tumor progression and metastasis, however, the potential association between FA-related genes (FARGs) and lung adenocarcinoma (LUAD) is still unclear. In this study, we described the genetic and transcriptomic changes of FARGs in LUAD patients and identified two different FA subtypes, which were significantly correlated with overall survival and tumor microenvironment infiltrating cells in LUAD patients. In addition, the FA score was also constructed through the LASSO Cox to evaluate the FA dysfunction of each patient. Multivariate Cox analysis proved that the FA score was an independent predictor and created the FA score integrated nomogram, which offered a quantitative tool for clinical practice. The performance of the FA score has been substantiated in numerous datasets for its commendable accuracy in estimating overall survival in LUAD patients. The groups with high and low FA scores exhibited different mutation spectrums, copy number variations, enrichment pathways, and immune status. Noteworthy differences between the two groups in terms of immunophenoscore and Tumor Immune Dysfunction and Exclusion were observed, suggesting that the group with a low FA score was more responsive to immunotherapy, and similar results were also confirmed in the immunotherapy cohort. In addition, seven potential chemotherapeutic drugs related to FA score targeting were predicted. Ultimately, we ascertained that the attenuation of KRT6A expression impeded the proliferation, migration, and invasion of LUAD cell lines. In summary, this research offers novel biomarkers to facilitate prognostic forecasting and clinical supervision for individuals afflicted with LUAD.
Collapse
Affiliation(s)
- Yuzhi Wang
- Department of Laboratory Medicine, Deyang People’s Hospital, Deyang 618000, Sichuan, People’s Republic of China
| | - Xiaoxiao Huang
- Department of Laboratory Medicine, Liuzhou Hospital of Guangzhou Women and Children’s Medical Center, Liuzhou 545000, Guangxi, People’s Republic of China
- Guangxi Clinical Research Center for Obstetrics and Gynecology, Liuzhou 545000, Guangxi, People’s Republic of China
| | - Hong Fan
- Department of Pathology, Shanghai Jianding District Anting Hospital, Shanghai 200000, People’s Republic of China
| | - Yunfei Xu
- Department of Laboratory Medicine, Chengdu Women’s and Children’s Central Hospital, Chengdu 610031, Sichuan, People’s Republic of China
| | - Zelin Qi
- Department of Laboratory Medicine, Deyang People’s Hospital, Deyang 618000, Sichuan, People’s Republic of China
| | - Yi Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, Fujian, People’s Republic of China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou 350001, Fujian, People’s Republic of China
| | - Yi Huang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, Fujian, People’s Republic of China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou 350001, Fujian, People’s Republic of China
- Central Laboratory, Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou 350001, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fuzhou 350001, Fujian, People’s Republic of China
| |
Collapse
|
30
|
Ohmoto A, Fuji S. Rapid T-cell lymphoma progression associated with immune checkpoint inhibitors. Expert Rev Hematol 2023:1-7. [PMID: 37191476 DOI: 10.1080/17474086.2023.2215424] [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: 05/17/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are widely used for multiple types of malignancies and are considered the fourth pillar in cancer treatment. Anti-programmed death-1 (PD-1) antibodies pembrolizumab and nivolumab are approved for relapsed/refractory classical Hodgkin lymphoma. Nonetheless, two phase 2 trials for T-cell lymphoma were terminated because of hyperprogression after a single dose in some patients. AREAS COVERED In this review, we summarize available information on the rapid progression of peripheral T-cell lymphoma including adult T-cell leukemia/lymphoma (ATLL). EXPERT OPINION In the abovementioned two trials, disease subtypes in patients who experienced hyperprogression were mostly ATLL or angioimmunoblastic T-cell lymphoma. Possible hyperprogression mechanisms induced by PD-1 blockade are the compensatory upregulation of the expression of other checkpoints, altered expression of lymphoma-promoting growth factors, functional blockade of stromal PD-ligand 1 acting as a tumor suppressor, and unique immune environment in indolent ATLL. The differentiation between hyperprogression and pseudoprogression is practically essential. There are no established methods to predict hyperprogression before administration of an ICI. In the future, the progress of novel diagnostic modalities such as positron emission tomography with computed tomography and circulating tumor DNA is expected to facilitate early cancer detection.
Collapse
Affiliation(s)
- Akihiro Ohmoto
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 1358550, Japan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shigeo Fuji
- Department of Hematology, Osaka International Cancer Institute, Osaka, 5418567, Japan
| |
Collapse
|
31
|
Ma X, Fang W, Wang D, Shao N, Chen J, Nie T, Huang C, Huang Y, Luo L, Xiao Z. Nanomaterial-Based Antivascular Therapy in the Multimodal Treatment of Cancer. Pharmaceutics 2023; 15:pharmaceutics15041207. [PMID: 37111692 PMCID: PMC10145863 DOI: 10.3390/pharmaceutics15041207] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Abnormal tumor vasculature and a hypoxic tumor microenvironment (TME) limit the effectiveness of conventional cancer treatment. Recent studies have shown that antivascular strategies that focus on antagonizing the hypoxic TME and promoting vessel normalization effectively synergize to increase the antitumor efficacy of conventional therapeutic regimens. By integrating multiple therapeutic agents, well-designed nanomaterials exhibit great advantages in achieving higher drug delivery efficiency and can be used as multimodal therapy with reduced systemic toxicity. In this review, strategies for the nanomaterial-based administration of antivascular therapy combined with other common tumor treatments, including immunotherapy, chemotherapy, phototherapy, radiotherapy, and interventional therapy, are summarized. In particular, the administration of intravascular therapy and other therapies with the use of versatile nanodrugs is also described. This review provides a reference for the development of multifunctional nanotheranostic platforms for effective antivascular therapy in combined anticancer treatments.
Collapse
Affiliation(s)
- Xiaocong Ma
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Weimin Fang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Duo Wang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Ni Shao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Jifeng Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Tianqi Nie
- The 12th People's Hospital of Guangzhou, Guangzhou 510620, China
| | - Cuiqing Huang
- Department of Ultrasound, Guangdong Women and Children Hospital, Guangzhou 511400, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Liangping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| |
Collapse
|
32
|
Cai Z, Chen J, Yu Z, Li H, Liu Z, Deng D, Liu J, Chen C, Zhang C, Ou Z, Chen M, Hu J, Zu X. BCAT2 Shapes a Noninflamed Tumor Microenvironment and Induces Resistance to Anti-PD-1/PD-L1 Immunotherapy by Negatively Regulating Proinflammatory Chemokines and Anticancer Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207155. [PMID: 36642843 PMCID: PMC10015882 DOI: 10.1002/advs.202207155] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Indexed: 06/17/2023]
Abstract
To improve response rate of monotherapy of immune checkpoint blockade (ICB), it is necessary to find an emerging target in combination therapy. Through analyzing tumor microenvironment (TME)-related indicators, it is validated that BCAT2 shapes a noninflamed TME in bladder cancer. The outcomes of multiomics indicate that BCAT2 has an inhibitory effect on cytotoxic lymphocyte recruitment by restraining activities of proinflammatory cytokine/chemokine-related pathways and T-cell-chemotaxis pathway. Immunoassays reveal that secretion of CD8+ T-cell-related chemokines keeps a robust negative correlation with BCAT2, generating a decreasing tendency of CD8+ T cells around BCAT2+ tumor cells from far to near. Cotreatment of BCAT2 deficiency and anti-PD-1 antibody has a synergistic effect in vivo, implying the potential of BCAT2 in combination therapy. Moreover, the value of BCAT2 in predicting efficacy of immunotherapy is validated in multiple immunotherapy cohorts. Together, as a key molecule in TME, BCAT2 is an emerging target in combination with ICB and a biomarker of guiding precision therapy.
Collapse
Affiliation(s)
- Zhiyong Cai
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jinbo Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zhengzheng Yu
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- Research Center of Carcinogenesis and Targeted TherapyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Huihuang Li
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zhi Liu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Dingshan Deng
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jinhui Liu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Chunliang Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Chunyu Zhang
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zhenyu Ou
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Minfeng Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jiao Hu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Xiongbing Zu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| |
Collapse
|
33
|
Jiang G, Miao Y, Wang Z, Zhang Q, Zhou P, Zhang F. Prognostic significance of epidermal growth factor receptor and programmed cell death-ligand 1 co-expression in esophageal squamous cell carcinoma. Aging (Albany NY) 2023; 15:1107-1129. [PMID: 36812484 PMCID: PMC10008495 DOI: 10.18632/aging.204535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023]
Abstract
Our study aimed to observe the correlation between epidermal growth factor receptor (EGFR) and programmed cell death-ligand 1 (PD-L1) expression and evaluate prognostic potential of their co-expression in esophageal squamous cell carcinoma (ESCC) patients. EGFR and PD-L1 expression were evaluated by immunohistochemical analysis. We revealed that there was a positive correlation between EGFR and PD-L1 expression in ESCC (P = 0.004). According to the positive relationship between EGFR and PD-L1, all patients were divided into four groups: EGFR (+)/PD-L1 (+), EGFR (+)/PD-L1 (-), EGFR (-)/PD-L1 (+), and EGFR (-)/PD-L1 (-). In 57 ESCC patients without surgery, we found that EGFR and PD-L1 co-expression were statistically correlated with a lower objective response rate (ORR) (p = 0.029), overall survival (OS) (p = 0.018) and progression-free survival (PFS) (p = 0.045) than those with one or none positive protein. Furthermore, PD-L1 expression has a significant positive correlation with infiltration level of 19 immune cells, EGFR expression was significantly correlated with infiltration level of 12 immune cells. The infiltration level of CD8 T cell and B cell were negatively correlated with EGFR expression. On the contrary with EGFR, the infiltration level of CD8 T cell, and B cell were positively correlated with PD-L1 expression. In conclusion, EGFR and PD-L1 co-expression could predict poor ORR and survival in ESCC without surgery, indicating a subset of patients who may benefit from a combination of targeted therapy against EGFR and PD-L1, which may expand the population benefiting from immunotherapy and reduce the occurrence of hyper progressive diseases.
Collapse
Affiliation(s)
- Guoxiang Jiang
- Department of Oncology Radiotherapy, Yantaishan Hospital, Yantai 264025, Shandong, China
| | - Yandong Miao
- Department of Oncology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264000, Shandong, China
| | - Zhenbo Wang
- Department of Radiation Oncology, Binzhou Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong, China
| | - Qi Zhang
- Department of Pathology, The First Hospital of Zibo, Zibo 255200, Shandong, China
| | - Ping Zhou
- Department of Pathology, The First Hospital of Zibo, Zibo 255200, Shandong, China
| | - Fang Zhang
- Department of Radiation Oncology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 26400, Shandong, China
| |
Collapse
|
34
|
Cai H, Ren Y, Chen S, Wang Y, Chu L. Ferroptosis and tumor immunotherapy: A promising combination therapy for tumors. Front Oncol 2023; 13:1119369. [PMID: 36845720 PMCID: PMC9945274 DOI: 10.3389/fonc.2023.1119369] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
Low response rate and treatment resistance are frequent problems in the immunotherapy of tumors, resulting in the unsatisfactory therapeutic effects. Ferroptosis is a form of cell death characterized by the accumulation of lipid peroxides. In recent years, it has been found that ferroptosis may be related to the treatment of cancer. Various immune cells (including macrophages and CD8+ T cells) can induce ferroptosis of tumor cells, and synergistically enhance the anti-tumor immune effects. However, the mechanisms are different for each cell types. DAMP released in vitro by cancer cells undergoing ferroptosis lead to the maturation of dendritic cells, cross-induction of CD8+ T cells, IFN-γ production and M1 macrophage production. Thus, it activates the adaptability of the tumor microenvironment and forms positive feedback of the immune response. It suggests that induction of ferroptosis may contribute to reducing resistance of cancer immunotherapy and has great potential in cancer therapy. Further research into the link between ferroptosis and tumor immunotherapy may offer hope for those cancers that are difficult to treat. In this review, we focus on the role of ferroptosis in tumor immunotherapy, explore the role of ferroptosis in various immune cells, and discuss potential applications of ferroptosis in tumor immunotherapy.
Collapse
Affiliation(s)
- Huazhong Cai
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China,*Correspondence: Huazhong Cai,
| | - Yongfei Ren
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Shuangwei Chen
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yue Wang
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Liangmei Chu
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
35
|
Zhu L, Zhang XP, Xu S, Hu MG, Zhao ZM, Zhao GD, Xiao ZH, Liu R. Identification of a CD4+ conventional T cells-related lncRNAs signature associated with hepatocellular carcinoma prognosis, therapy, and tumor microenvironment. Front Immunol 2023; 13:1111246. [PMID: 36700197 PMCID: PMC9868629 DOI: 10.3389/fimmu.2022.1111246] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death worldwide, and CD4+ T lymphocytes can inhibit hepatocarcinogenesis and mediate tumor regression. However, few studies have focused on the prognostic power of CD4+ Tconv-related lncRNAs in HCC patients. Method We obtained data from TCGA and GEO databases and identified CD4+Tconv-related lncRNAs in HCC. The risk score was constructed using lasso regression and the model was validated using two validation cohorts. The RS was also assessed in different clinical subgroups, and a nomogram was established to further predict the patients' outcomes. Furthermore, we estimated the immune cell infiltration and cancer-associated fibroblasts (CAFs) through TIMER databases and assessed the role of RS in immune checkpoint inhibitors response. Results We constructed a CD4+ Tconv-related lncRNAs risk score, including six lncRNAs (AC012073.1, AL031985.3, LINC01060, MKLN1-AS, MSC-AS1, and TMCC1-AS1), and the RS had good predictive ability in validation cohorts and most clinical subgroups. The RS and the T stage were included in the nomogram with optimum prediction and the model had comparable OS prediction power compared to the AJCC. Patients in the high-risk group had a poor immune response phenotype, with high infiltrations of macrophages, CAFs, and low infiltrations of NK cells. Immunotherapy and chemotherapy response analysis indicated that low-risk group patients had good reactions to immune checkpoint inhibitors. Conclusion We constructed and validated a novel CD4+ Tconv-related lncRNAs RS, with the potential predictive value of HCC patients' survival and immunotherapy response.
Collapse
Affiliation(s)
- Lin Zhu
- Medical School of Chinese PLA, Beijing, China,Faculty of Hepato-Biliary-Pancreatic Surgery, the First Medical Centre, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China,Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China,Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China,The First Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xiu-Ping Zhang
- Medical School of Chinese PLA, Beijing, China,Faculty of Hepato-Biliary-Pancreatic Surgery, the First Medical Centre, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China,Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China,Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China
| | - Shuai Xu
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ming-Gen Hu
- Faculty of Hepato-Biliary-Pancreatic Surgery, the First Medical Centre, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China,Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China,Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China
| | - Zhi-Ming Zhao
- Faculty of Hepato-Biliary-Pancreatic Surgery, the First Medical Centre, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China,Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China,Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China
| | - Guo-Dong Zhao
- Faculty of Hepato-Biliary-Pancreatic Surgery, the First Medical Centre, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China,Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China,Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China
| | - Zhao-Hui Xiao
- Faculty of Hepato-Biliary-Pancreatic Surgery, the First Medical Centre, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China,Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China,Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China
| | - Rong Liu
- Medical School of Chinese PLA, Beijing, China,Faculty of Hepato-Biliary-Pancreatic Surgery, the First Medical Centre, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China,Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China,Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China,The First Clinical Medical School, Lanzhou University, Lanzhou, China,*Correspondence: Rong Liu,
| |
Collapse
|
36
|
Wang X, Guo Z, Wu X, Chen D, Wang F, Yang L, Luo M, Wu S, Yang C, Huang L, Fu L. Predictive Nomogram for Hyperprogressive Disease During Anti-PD-1/PD-L1 Treatment in Patients with Advanced Non-Small Cell Lung Cancer. Immunotargets Ther 2023; 12:1-16. [PMID: 36632330 PMCID: PMC9828302 DOI: 10.2147/itt.s373866] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/29/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction Various studies have reported that anti-PD-1/PD-L1 treatment may lead to the rapid development of tumors called hyperprogressive disease (HPD). A nomogram for HPD prediction in NSCLC patients is urgently needed. Methods This retrospective cohort study included 176 cases for establishing a model of HPD prediction and 85 cases for validation in advanced NSCLC patients treated with PD-1/PD-L1 inhibitors. HPD was defined as tumor growth rate (TGR, ≥ 2), tumor growth kinetics (TGK, ≥ 2) or time to treatment failure (TTF, ≤ 2 months). Univariate and multivariate logistic regression were used to estimate the specified factors associated with HPD. Then, the nomogram was developed and validated. Results Anti-PD-1/PD-L1 therapy resulted in a 9.66% (17/176) incidence of HPD in advanced NSCLC. The overall survival (OS) and progression-free survival (PFS) in patients with HPD were significantly shorter than those in patients without HPD (OS: 7.00 vs 12.00 months, P<0.01; PFS: 2.00 vs 5.00 months, P<0.001, respectively). The HPD prediction nomogram included APTT (P<0.01), CD4+ CD25+ CD127-low cells (Treg cells) (P<0.01), the presence of liver metastasis (P<0.05), and more than two metastatic sites (P<0.05). Then, patients were divided into two groups by the "HPD score" calculated by the nomogram. The C-index was 0.845, while the area under the curve (AUC) was 0.830 (sensitivity 75.00%, specificity 91.70%). The calibration plot of HPD probability showed an optimal agreement between the actual observation and prediction by the nomogram. In the validation cohort, the AUC was up to 0.960 (sensitivity 88.70%, specificity 89.80%). Conclusions The nomogram was constructed with the presence of liver metastasis, more than two metastatic sites, lengthened APTT and a high level of Treg cells, which could be used to predict HPD risk.
Collapse
Affiliation(s)
- Xueping Wang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, Guangzhou, 510060, People’s Republic of China
| | - Zhixing Guo
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, Guangzhou, 510060, People’s Republic of China
| | - Xingping Wu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, Guangzhou, 510060, People’s Republic of China
| | - Da Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, Guangzhou, 510060, People’s Republic of China
| | - Fang Wang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, Guangzhou, 510060, People’s Republic of China
| | - Lewei Yang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, Guangzhou, 510060, People’s Republic of China
| | - Min Luo
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, Guangzhou, 510060, People’s Republic of China
| | - Shaocong Wu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, Guangzhou, 510060, People’s Republic of China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, Guangzhou, 510060, People’s Republic of China
| | - Lamei Huang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, Guangzhou, 510060, People’s Republic of China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, Guangzhou, 510060, People’s Republic of China,Correspondence: Liwu Fu, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Cancer Center, Sun Yat-sen University, 651 Dongfengdong Road, Guangzhou, 510060, People’s Republic of China, Email
| |
Collapse
|
37
|
Zheng LP, Yang J, Chen XW, Li LC, Sun JG. Correlation of preclinical and clinical biomarkers with efficacy and toxicity of cancer immunotherapy. Ther Adv Med Oncol 2023; 15:17588359231163807. [PMID: 37113734 PMCID: PMC10126660 DOI: 10.1177/17588359231163807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 02/27/2023] [Indexed: 04/29/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revealed significant clinical values in different solid tumors and hematological malignancy, changing the landscape for the treatment of multiple types of cancer. However, only a subpopulation of patients has obvious tumor response and long-term survival after ICIs treatment, and many patients may experience other undesirable clinical features. Therefore, biomarkers are critical for patients to choose exact optimum therapy. Here, we reviewed existing preclinical and clinical biomarkers of immunotherapeutic efficacy and immune-related adverse events (irAEs). Based on efficacy prediction, pseudoprogression, hyperprogressive disease, or irAEs, these biomarkers were divided into cancer cell-derived biomarkers, tumor microenvironment-derived biomarkers, host-derived biomarkers, peripheral blood biomarkers, and multi-modal model and artificial intelligence assessment-based biomarkers. Furthermore, we describe the relation between ICIs efficacy and irAEs. This review provides the overall perspective of biomarkers of immunotherapeutic outcome and irAEs prediction during ICIs treatment.
Collapse
Affiliation(s)
| | | | - Xie-Wan Chen
- Department of Basic Medicine, Army Medical University, Chongqing, China
| | - Ling-Chen Li
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | | |
Collapse
|
38
|
Zhang Y, Wu J, Zhao C, Zhang S, Zhu J. Recent Advancement of PD-L1 Detection Technologies and Clinical Applications in the Era of Precision Cancer Therapy. J Cancer 2023; 14:850-873. [PMID: 37056391 PMCID: PMC10088895 DOI: 10.7150/jca.81899] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/14/2023] [Indexed: 04/15/2023] Open
Abstract
Programmed death-1 is a protein found on the surface of immune cells that can interact with its ligand, programmed death-ligand 1 (PD-L1), which is expressed on the plasma membrane, the surface of secreted cellular exosomes, in cell nuclei, or as a circulating soluble protein. This interaction can lead to immune escape in cancer patients. In clinical settings, PD-L1 plays an important role in tumor disease diagnosis, determining therapeutic effectiveness, and predicting patient prognosis. PD-L1 inhibitors are also essential components of tumor immunotherapy. Thus, the detection of PD-L1 levels is crucial, especially in the era of precision cancer therapy. In recent years, innovations have been made in traditional immunoassay methods and the development of new immunoassays for PD-L1 detection. This review aims to summarize recent research progress in tumor PD-L1 detection technology and highlight the clinical applications of PD-L1.
Collapse
Affiliation(s)
- Yuanfeng Zhang
- Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Juanjuan Wu
- Binzhou People's Hospital Affiliated to Shandong First Medical University, Binzhou, Shandong, 256600, China
| | - Chaobin Zhao
- Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Shuyuan Zhang
- Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Jianbo Zhu
- Binzhou People's Hospital Affiliated to Shandong First Medical University, Binzhou, Shandong, 256600, China
- ✉ Corresponding author: Pro. Jianbo Zhu, Binzhou People's Hospital Affiliated to Shandong First Medical University, 515 Yellow River Seven Road, Binzhou, Shandong, 256600, China; ,
| |
Collapse
|
39
|
Lin G, Wang X, Ye H, Cao W. Radiomic Models Predict Tumor Microenvironment Using Artificial Intelligence-the Novel Biomarkers in Breast Cancer Immune Microenvironment. Technol Cancer Res Treat 2023; 22:15330338231218227. [PMID: 38111330 PMCID: PMC10734346 DOI: 10.1177/15330338231218227] [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: 09/12/2023] [Revised: 10/22/2023] [Accepted: 11/16/2023] [Indexed: 12/20/2023] Open
Abstract
Breast cancer is the most common malignancy in women, and some subtypes are associated with a poor prognosis with a lack of efficacious therapy. Moreover, immunotherapy and the use of other novel antibody‒drug conjugates have been rapidly incorporated into the standard management of advanced breast cancer. To extract more benefit from these therapies, clarifying and monitoring the tumor microenvironment (TME) status is critical, but this is difficult to accomplish based on conventional approaches. Radiomics is a method wherein radiological image features are comprehensively collected and assessed to build connections with disease diagnosis, prognosis, therapy efficacy, the TME, etc In recent years, studies focused on predicting the TME using radiomics have increasingly emerged, most of which demonstrate meaningful results and show better capability than conventional methods in some aspects. Beyond predicting tumor-infiltrating lymphocytes, immunophenotypes, cytokines, infiltrating inflammatory factors, and other stromal components, radiomic models have the potential to provide a completely new approach to deciphering the TME and facilitating tumor management by physicians.
Collapse
Affiliation(s)
- Guang Lin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiaojia Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Hunan Ye
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wenming Cao
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
40
|
Kanjanapan Y, Guduguntla G, Varikara AK, Szajer J, Yip D, Cockburn J, Fadia M. Hyperprogressive Disease (HPD) in Solid Tumours Receiving Immune Checkpoint Inhibitors in a Real-World Setting. Technol Cancer Res Treat 2023; 22:15330338231209129. [PMID: 37885403 PMCID: PMC10612441 DOI: 10.1177/15330338231209129] [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: 06/15/2023] [Revised: 09/15/2023] [Accepted: 10/04/2023] [Indexed: 10/28/2023] Open
Abstract
Introduction: Hyperprogressive disease (HPD) is a state of accelerated tumor growth from cancer immunotherapy, associated with poor outcome. The reported incidence is 6% to 29% among studies using varying definitions of HPD, with no predictive biomarkers. Tumor infiltrating lymphocytes (TILs) are prognostic and predictive for immunotherapy benefit in various tumor types, but have only been tested for correlation with HPD in one study. Objectives: The objective of the study was to determine the prevalence of HPD in solid tumor patients treated with immune checkpoint inhibitor therapy in a real-world setting, and to assess clinicopathological features as potential biomarkers for HPD. Methods: We conducted a retrospective analysis of solid tumor patients treated with immune checkpoint inhibitors at a single institution. Imaging pre-immunotherapy and postimmunotherapy were assessed for HPD, and correlated against clinicopathological factors, including TILs and programmed death-ligand 1 (PD-L1) status through archival tumor assessment. HPD was defined per Matos et al as response evaluation criteria in solid tumors (RECIST) progressive disease, minimum increase in measurable lesions of 10 mm, plus increase of ≥40% in sum of target lesions compared with baseline and/or increase of ≥20% in sum of target lesions compared with baseline plus new lesions in at least 2 different organs. Results: HPD occurred in 11 of 87 patients (13%), and associated with inferior overall survival (median 5.5 months vs 18.3 months, P = .002). However, on multivariate analysis, only liver metastases (hazard ratio [HR] 4.66, 95% confidence interval [CI] 2.27-9.56, P < .001) and PD-L1 status (HR 0.53, 95% CI 0.30-0.95, P = .03) were significantly associated with survival. Presence of liver metastases correlated with occurence of HPD (P = .01). Age, sex, and monotherapy versus combination immunotherapy were not predictive for HPD. PD-L1 status and TILs were not associated with HPD. Conclusions: We found 13% HPD among solid tumor patients treated with immunotherapy, consistent with the range reported in prior series. Assessment for HPD is feasible outside of a clinical trials setting, using modified criteria that require comparison of 2 imaging studies. Liver metastases were associated with risk of HPD, while TILs and PD-L1 status were not predictive for HPD.
Collapse
Affiliation(s)
- Yada Kanjanapan
- Department of Medical Oncology, The Canberra Hospital, Canberra, Australia
- ANU Medical School, Australian National University, Canberra, Australia
| | - Geetha Guduguntla
- Department of Medical Imaging, The Canberra Hospital, Canberra, Australia
| | | | - Jeremy Szajer
- Department of Medical Imaging, The Canberra Hospital, Canberra, Australia
| | - Desmond Yip
- Department of Medical Oncology, The Canberra Hospital, Canberra, Australia
- ANU Medical School, Australian National University, Canberra, Australia
| | - John Cockburn
- ANU Medical School, Australian National University, Canberra, Australia
- Department of Medical Imaging, The Canberra Hospital, Canberra, Australia
| | - Mitali Fadia
- ANU Medical School, Australian National University, Canberra, Australia
- Department of Pathology, The Canberra Hospital, Canberra, Australia
| |
Collapse
|
41
|
Fan J, To KKW, Chen ZS, Fu L. ABC transporters affects tumor immune microenvironment to regulate cancer immunotherapy and multidrug resistance. Drug Resist Updat 2023; 66:100905. [PMID: 36463807 DOI: 10.1016/j.drup.2022.100905] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/16/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022]
Abstract
Multidrug resistance (MDR) is the phenomenon in which cancer cells simultaneously develop resistance to a broad spectrum of structurally and mechanistically unrelated drugs. MDR severely hinders the effective treatment of cancer and is the major cause of chemotherapy failure. ATP-binding cassette (ABC) transporters are extensively expressed in various body tissues, and actively transport endogenous and exogenous substrates through biological membranes. Overexpression of ABC transporters is frequently observed in MDR cancer cells, which promotes efflux of chemotherapeutic drugs and reduces their intracellular accumulation. Increasing evidence suggests that ABC transporters regulate tumor immune microenvironment (TIME) by transporting various cytokines, thus controlling anti-tumor immunity and sensitivity to anticancer drugs. On the other hand, the expression of various ABC transporters is regulated by cytokines and other immune signaling molecules. Targeted inhibition of ABC transporter expression or function can enhance the efficacy of immune checkpoint inhibitors by promoting anticancer immune microenvironment. This review provides an update on the recent research progress in this field.
Collapse
Affiliation(s)
- Jingyi Fan
- State Key Laboratory of Oncology in South China;Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China; Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing 100038, China
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States.
| | - Liwu Fu
- State Key Laboratory of Oncology in South China;Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
42
|
Fu K, Xie F, Wang F, Fu L. Therapeutic strategies for EGFR-mutated non-small cell lung cancer patients with osimertinib resistance. J Hematol Oncol 2022; 15:173. [PMID: 36482474 PMCID: PMC9733018 DOI: 10.1186/s13045-022-01391-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/26/2022] [Indexed: 12/13/2022] Open
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are the preferential options for advanced non-small cell lung cancer (NSCLC) patients harboring EGFR mutations. Osimertinib is a potent irreversible third-generation EGFR-TKI targeting EGFR mutations but has little effect on wild-type EGFR. In view of its remarkable efficacy and manageable safety, osimertinib was recommended as the standard first-line treatment for advanced or metastatic NSCLC patients with EGFR mutations. However, as the other EGFR-TKIs, osimertinib will inevitably develop acquired resistance, which limits its efficacy on the treatment of EGFR-mutated NSCLC patients. The etiology of triggering osimertinib resistance is complex including EGFR-dependent and EGFR-independent pathways, and different therapeutic strategies for the NSCLC patients with osimertinib resistance have been developed. Herein, we comprehensively summarized the resistance mechanisms of osimertinib and discuss in detail the potential therapeutic strategies for EGFR-mutated NSCLC patients suffering osimertinib resistance for the sake of the improvement of survival and further achievement of precise medicine.
Collapse
Affiliation(s)
- Kai Fu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Fachao Xie
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Fang Wang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Liwu Fu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| |
Collapse
|
43
|
Pawłowska A, Skiba W, Suszczyk D, Kuryło W, Jakubowicz-Gil J, Paduch R, Wertel I. The Dual Blockade of the TIGIT and PD-1/PD-L1 Pathway as a New Hope for Ovarian Cancer Patients. Cancers (Basel) 2022; 14:5757. [PMID: 36497240 PMCID: PMC9740841 DOI: 10.3390/cancers14235757] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
The prognosis for ovarian cancer (OC) patients is poor and the five-year survival rate is only 47%. Immune checkpoints (ICPs) appear to be the potential targets in up-and-coming OC treatment. However, the response of OC patients to immunotherapy based on programmed cell death pathway (PD-1/PD-L1) inhibitors totals only 6-15%. The promising approach is a combined therapy, including other ICPs such as the T-cell immunoglobulin and ITIM domain/CD155/DNAX accessory molecule-1 (TIGIT/CD155/DNAM-1) axis. Preclinical studies in a murine model of colorectal cancer showed that the dual blockade of PD-1/PD-L1 and TIGIT led to remission in the whole studied group vs. the regression of the tumors with the blockade of a single pathway. The approach stimulates the effector activity of T cells and NK cells, and redirects the immune system activity against the tumor. The understanding of the synergistic action of the TIGIT and PD-1/PD-L1 blockade is, however, poor. Thus, the aim of this review is to summarize the current knowledge about the mode of action of the dual TIGIT and PD-1/PD-L1 blockade and its potential benefits for OC patients. Considering the positive impact of this combined therapy in malignancies, including lung and colorectal cancer, it appears to be a promising approach in OC treatment.
Collapse
Affiliation(s)
- Anna Pawłowska
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Wiktoria Skiba
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Dorota Suszczyk
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Weronika Kuryło
- Students’ Scientific Association, Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Joanna Jakubowicz-Gil
- Department of Functional Anatomy and Cytobiology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Roman Paduch
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| |
Collapse
|
44
|
Harkus U, Wankell M, Palamuthusingam P, McFarlane C, Hebbard L. Immune checkpoint inhibitors in HCC: Cellular, molecular and systemic data. Semin Cancer Biol 2022; 86:799-815. [PMID: 35065242 DOI: 10.1016/j.semcancer.2022.01.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 01/27/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer related deaths in the world, and for patients with advanced disease there are few therapeutic options available. The complex immunological microenvironment of HCC and the success of immunotherapy in several types of tumours, has raised the prospect of potential benefit for immune based therapies, such as immune checkpoint inhibitors (ICIs), in HCC. This has led to significant breakthrough research, numerous clinical trials and the rapid approval of multiple systemic drugs for HCC by regulatory bodies worldwide. Although some patients responded well to ICIs, many have failed to achieve significant benefit, while others showed unexpected and paradoxical deterioration. The aim of this review is to discuss the pathophysiology of HCC, the tumour microenvironment, key clinical trials evaluating ICIs in HCC, various resistance mechanisms to ICIs, and possible ways to overcome these impediments to improve patient outcomes.
Collapse
Affiliation(s)
- Uasim Harkus
- Townsville University Hospital, Townsville, Queensland 4811, Australia
| | - Miriam Wankell
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland 4811, Australia
| | - Pranavan Palamuthusingam
- College of Medicine and Dentistry, James Cook University, Townsville, Queensland 4811, Australia; Townsville University Hospital, Townsville, Queensland 4811, Australia; Mater Hospital, Townsville, Queensland 4811, Australia
| | - Craig McFarlane
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland 4811, Australia
| | - Lionel Hebbard
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland 4811, Australia; Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales 2145, Australia.
| |
Collapse
|
45
|
Ali A, Davidson S, Fraenkel E, Gilmore I, Hankemeier T, Kirwan JA, Lane AN, Lanekoff I, Larion M, McCall LI, Murphy M, Sweedler JV, Zhu C. Single cell metabolism: current and future trends. Metabolomics 2022; 18:77. [PMID: 36181583 PMCID: PMC10063251 DOI: 10.1007/s11306-022-01934-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022]
Abstract
Single cell metabolomics is an emerging and rapidly developing field that complements developments in single cell analysis by genomics and proteomics. Major goals include mapping and quantifying the metabolome in sufficient detail to provide useful information about cellular function in highly heterogeneous systems such as tissue, ultimately with spatial resolution at the individual cell level. The chemical diversity and dynamic range of metabolites poses particular challenges for detection, identification and quantification. In this review we discuss both significant technical issues of measurement and interpretation, and progress toward addressing them, with recent examples from diverse biological systems. We provide a framework for further directions aimed at improving workflow and robustness so that such analyses may become commonly applied, especially in combination with metabolic imaging and single cell transcriptomics and proteomics.
Collapse
Affiliation(s)
- Ahmed Ali
- Leiden Academic Centre for Drug Research, University of Leiden, Gorlaeus Building Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Shawn Davidson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Ernest Fraenkel
- Department of Biological Engineering and the Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ian Gilmore
- National Physical Laboratory, Teddington, TW11 0LW, Middlesex, UK
| | - Thomas Hankemeier
- Leiden Academic Centre for Drug Research, University of Leiden, Room number GW4.07, Gorlaeus Building, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Jennifer A Kirwan
- Berlin Institute of Health, Metabolomics Platform, Translational Research Unit of the Charite-Universitätsmedizin Berlin, Anna-Louisa-Karsch-Str 2, 10178, Berlin, Germany
| | - Andrew N Lane
- Department of Toxicology and Cancer Biology, and Center for Environmental and Systems Biochemistry, University of Kentucky, 789 S. Limestone St, Lexington, KY, 40536, USA.
| | - Ingela Lanekoff
- Department of Chemistry-BMC, Uppsala University, Husargatan 3 (576), 751 23, Uppsala, Sweden
| | - Mioara Larion
- Center for Cancer Research, National Cancer Institute, Building 37, Room 1136A, Bethesda, MD, 20892, USA
| | - Laura-Isobel McCall
- Department of Chemistry & Biochemistry, Department of Microbiology and Plant Biology, Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, 101 Stephenson Parkway, room 3750, Norman, OK, 73019-5251, USA
| | - Michael Murphy
- Departments of Biological Engineering, Department of Electrical Engineering, and Computer Science and the Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, USA
| | - Jonathan V Sweedler
- Department of Chemistry, and the Beckman Institute, University of Illinois Urbana-Champaign, 505 South Mathews Avenue, Urbana, IL, 61801, USA
| | - Caigang Zhu
- Department of Biomedical Engineering, University of Kentucky, Lexington, KY, 40536, USA
| |
Collapse
|
46
|
Walters MK, Ackerman AT, Weese JL, Ruggeri A, Mullane MP, Hunt A, Wilson A, Ramczyk BL, Thompson MA. Quantifying the Value of the Molecular Tumor Board: Discordance Recommendation Rate and Drug Cost Avoidance. JCO Precis Oncol 2022; 6:e2200132. [PMID: 36265115 DOI: 10.1200/po.22.00132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/16/2022] [Accepted: 08/31/2022] [Indexed: 06/16/2023] Open
Abstract
PURPOSE Molecular tumor boards (MTBs) provide interventions that assist the patient's primary oncologist's interpretation and application of precision oncology and avoid clinical and financial toxicities of prescribing inappropriate targeted therapy. In this article, we describe a novel method for illustrating MTBs value and recommendation discordance rate and report associated drug cost avoidance data. METHODS From January 1, 2021, to December 31, 2021, patients assessed by our program's MTB were retrospectively evaluated. Recommendation discordance was defined as any disagreement between MTB therapeutic recommendations and those provided in the next-generation sequencing vendor's report. RESULTS In 2021, our program processed 1,119 next-generation sequencing orders via external vendors for 1,029 unique patients with a variety of solid tumor and hematologic malignancies. During this period, 962 patients were reviewed through our MTB process. MTB recommendation discordance rate was high (229 of 502; 45.6%) and varied across test vendors. Rationales for discordance included the following: low level of evidence (88% of patients), alternative standard of care available (60%), and tolerability concerns (42%), among others. Discordance was highest for Vendor C (30%), followed by Vendor A (24%) and Vendor B (8%). The most common drug classes not supported were mTOR, PARP, MEK, and PIK3CA inhibitors when recommended by vendors in off-label settings. MTB interventions accounted for $3,209,070 in US dollars in potential drug cost avoidance. CONCLUSION Therapeutic recommendation discordance rates can provide quantitative insight into the benefit of MTB. Discordance-associated drug cost avoidance further demonstrates MTB's financial value. These measures may be used as part of the justification for this service line within a cancer care program.
Collapse
Affiliation(s)
- Mary K Walters
- Aurora Cancer Care, Oncology Precision Medicine, Advocate Aurora Health, Milwaukee, WI
| | - Andrew T Ackerman
- Aurora Cancer Care, Oncology Precision Medicine, Advocate Aurora Health, Milwaukee, WI
| | - James L Weese
- Aurora Cancer Care, Oncology Precision Medicine, Advocate Aurora Health, Milwaukee, WI
| | - Antony Ruggeri
- Aurora Cancer Care, Oncology Precision Medicine, Advocate Aurora Health, Milwaukee, WI
| | - Michael P Mullane
- Aurora Cancer Care, Oncology Precision Medicine, Advocate Aurora Health, Milwaukee, WI
- Hereditary Cancer Prevention and Management Center, Advocate Aurora Health, Milwaukee, WI
| | - Alicia Hunt
- ACL Laboratories, Advocate Aurora Health, West Allis, WI
| | - Amanda Wilson
- ACL Laboratories, Advocate Aurora Health, West Allis, WI
| | - Brenda L Ramczyk
- Aurora Cancer Care, Oncology Precision Medicine, Advocate Aurora Health, Milwaukee, WI
- Hereditary Cancer Prevention and Management Center, Advocate Aurora Health, Milwaukee, WI
| | - Michael A Thompson
- Aurora Cancer Care, Oncology Precision Medicine, Advocate Aurora Health, Milwaukee, WI
- Tempus Labs, Chicago, IL
| |
Collapse
|
47
|
Liu K, Sun Q, Liu Q, Li H, Zhang W, Sun C. Focus on immune checkpoint PD-1/PD-L1 pathway: New advances of polyphenol phytochemicals in tumor immunotherapy. Biomed Pharmacother 2022; 154:113618. [DOI: 10.1016/j.biopha.2022.113618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/19/2022] [Accepted: 08/27/2022] [Indexed: 11/02/2022] Open
|
48
|
Therapeutic targets and biomarkers of tumor immunotherapy: response versus non-response. Signal Transduct Target Ther 2022; 7:331. [PMID: 36123348 PMCID: PMC9485144 DOI: 10.1038/s41392-022-01136-2] [Citation(s) in RCA: 159] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/25/2022] [Accepted: 07/25/2022] [Indexed: 02/05/2023] Open
Abstract
Cancers are highly complex diseases that are characterized by not only the overgrowth of malignant cells but also an altered immune response. The inhibition and reprogramming of the immune system play critical roles in tumor initiation and progression. Immunotherapy aims to reactivate antitumor immune cells and overcome the immune escape mechanisms of tumors. Represented by immune checkpoint blockade and adoptive cell transfer, tumor immunotherapy has seen tremendous success in the clinic, with the capability to induce long-term regression of some tumors that are refractory to all other treatments. Among them, immune checkpoint blocking therapy, represented by PD-1/PD-L1 inhibitors (nivolumab) and CTLA-4 inhibitors (ipilimumab), has shown encouraging therapeutic effects in the treatment of various malignant tumors, such as non-small cell lung cancer (NSCLC) and melanoma. In addition, with the advent of CAR-T, CAR-M and other novel immunotherapy methods, immunotherapy has entered a new era. At present, evidence indicates that the combination of multiple immunotherapy methods may be one way to improve the therapeutic effect. However, the overall clinical response rate of tumor immunotherapy still needs improvement, which warrants the development of novel therapeutic designs as well as the discovery of biomarkers that can guide the prescription of these agents. Learning from the past success and failure of both clinical and basic research is critical for the rational design of studies in the future. In this article, we describe the efforts to manipulate the immune system against cancer and discuss different targets and cell types that can be exploited to promote the antitumor immune response.
Collapse
|
49
|
Chen JS, Hsieh YC, Chou CH, Wu YH, Yang MH, Chu SH, Chao YS, Chen CN. Chidamide plus Tyrosine Kinase Inhibitor Remodel the Tumor Immune Microenvironment and Reduce Tumor Progression When Combined with Immune Checkpoint Inhibitor in Naïve and Anti-PD-1 Resistant CT26-Bearing Mice. Int J Mol Sci 2022; 23:10677. [PMID: 36142591 PMCID: PMC9504159 DOI: 10.3390/ijms231810677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 11/25/2022] Open
Abstract
Combined inhibition of vascular endothelial growth factor receptor (VEGFR) and the programmed cell death protein 1 (PD-1) pathways has shown efficacy in multiple cancers; however, the clinical outcomes show limited benefits and the unmet clinical needs still remain and require improvement in efficacy. Using murine colon carcinoma (CT26) allograft models, we examined the efficacy and elucidated novel tumor microenvironment (TME) remodeling mechanisms underlying the combination of chidamide (a benzamide-based class l histone deacetylase inhibitor; brand name in Taiwan, Kepida®) with VEGF receptor tyrosine kinase inhibitor (TKIs; cabozantinib/regorafenib, etc.) and immune checkpoint inhibitors (ICIs; anti-PD-1/anti-PD-L1/anti-CTLA-4 antibodies). The TME was assessed using flow cytometry and RNA-sequencing to determine the novel mechanisms and their correlation with therapeutic effects in mice with significant treatment response. Compared with ICI alone or cabozantinib/regorafenib + ICI, combination of chidamide + cabozantinib/regorafenib + ICI increased the tumor response and survival benefits. In particular, treatment of CT26-bearing mice with chidamide + regorafenib + anti-PD-1 antibody showed a better objective response rate (ORR) and overall survival (OS). Similar results were observed in anti-PD-1 treatment-resistant mice. After treatment with this optimal combination, in the TME, RNA-sequencing revealed that downregulated mRNAs were correlated with leukocyte migration, cell chemotaxis, and macrophage gene sets, and flow cytometry analysis showed that the cell numbers of myeloid-derived polymorphonuclear suppressor cells and tumor-associated macrophages were decreased. Accordingly, chidamide + regorafenib + anti-PD-1 antibody combination therapy could trigger a novel TME remodeling mechanism by attenuating immunosuppressive cells, and restoring T-cell activation to enhance ORR and OS. Our studies also showed that the addition of Chidamide to the regorafenib + anti-PD-1 Ab combination could induce a durable tumor-specific response by attenuating immune suppression in the TME. In addition, this result suggests that TME remodeling, mediated by epigenetic immunomodulator combined with TKI and ICI, would be more advantageous for achieving a high objective response rate, when compared to TKI plus ICI or ICI alone, and maintaining long-lasting antitumor activity.
Collapse
Affiliation(s)
- Jia-Shiong Chen
- New Drug Research and Development Center, Great Novel Therapeutics Biotech & Medicals Corporation (GNTbm), Taipei 100, Taiwan
| | | | - Cheng-Han Chou
- Department of Biology, Great Novel Therapeutics Biotech & Medicals Corporation (GNTbm), Taipei 100, Taiwan
| | - Yi-Hong Wu
- Department of Biology, Great Novel Therapeutics Biotech & Medicals Corporation (GNTbm), Taipei 100, Taiwan
| | - Mu-Hsuan Yang
- Department of Chemistry, Great Novel Therapeutics Biotech & Medicals Corporation (GNTbm), Taipei 100, Taiwan
| | - Sz-Hao Chu
- Department of Chemistry, Great Novel Therapeutics Biotech & Medicals Corporation (GNTbm), Taipei 100, Taiwan
| | - Ye-Su Chao
- New Drug Research and Development Center, Great Novel Therapeutics Biotech & Medicals Corporation (GNTbm), Taipei 100, Taiwan
| | - Chia-Nan Chen
- New Drug Research and Development Center, Great Novel Therapeutics Biotech & Medicals Corporation (GNTbm), Taipei 100, Taiwan
| |
Collapse
|
50
|
Zhang Y, Li C, Xia C, Wah To KK, Guo Z, Ren C, Wen L, Wang F, Fu L, Liao N. Adagrasib, a KRAS G12C inhibitor, reverses the multidrug resistance mediated by ABCB1 in vitro and in vivo. Cell Commun Signal 2022; 20:142. [PMID: 36104708 PMCID: PMC9472360 DOI: 10.1186/s12964-022-00955-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/03/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Multidrug resistance (MDR) is a complex phenomenon that frequently leads to chemotherapy failure during cancer treatment. The overexpression of ATP-binding cassette (ABC) transporters represents the major mechanism contributing to MDR. To date, no effective MDR modulator has been applied in clinic. Adagrasib (MRTX849), a specific inhibitor targeting KRAS G12C mutant, is currently under investigation in clinical trials for the treatment of non-small cell lung cancer (NSCLC). This study focused on investigating the circumvention of MDR by MRTX849.
Methods
The cytotoxicity and MDR reversal effect of MRTX849 were assessed by MTT assay. Drug accumulation and drug efflux were evaluated by flow cytometry. The MDR reversal by MRTX849 in vivo was investigated in two ABCB1-overexpressing tumor xenograft models in nude mice. The interaction between MRTX849 and ABCB1 substrate binding sites was studied by the [125I]-IAAP-photoaffinity labeling assay. The vanadate-sensitive ATPase assay was performed to identify whether MRTX849 would change ABCB1 ATPase activity. The effect of MRTX849 on expression of ABCB1 and PI3K/AKT signaling molecules was examined by flow cytometry, Western blot and Quantitative Real-time PCR analyses.
Results
MRTX849 was shown to enhance the anticancer efficacy of ABCB1 substrate drugs in the transporter-overexpressing cells both in vitro and in vivo. The MDR reversal effect was specific against ABCB1 because no similar effect was observed in the parental sensitive cells or in ABCG2-mediated MDR cells. Mechanistically, MRTX849 increased the cellular accumulation of ABCB1 substrates including doxorubicin (Dox) and rhodamine 123 (Rho123) in ABCB1-overexpressing MDR cells by suppressing ABCB1 efflux activity. Additionally, MRTX849 stimulated ABCB1 ATPase activity and competed with [125I]-IAAP for photolabeling of ABCB1 in a concentration-dependent manner. However, MRTX849 did not alter ABCB1 expression or phosphorylation of AKT/ERK at the effective MDR reversal drug concentrations.
Conclusions
In summary, MRTX849 was found to overcome ABCB1-mediated MDR both in vitro and in vivo by specifically attenuating ABCB1 efflux activity in drug-resistant cancer cells. Further studies are warranted to translate the combination of MRTX849 and conventional chemotherapy to clinical application for circumvention of MDR.
Collapse
|