1
|
Fu R, Zhang C, Song MM, Gao X, Li F, Cai M, Jiang BY, Yang XN, Wu YL, Zhong WZ. A single-cell map of patients with non-small cell lung cancer harboring rare-driver mutations after anti-PD-1 treatment. Cancer Lett 2025; 616:217595. [PMID: 40021042 DOI: 10.1016/j.canlet.2025.217595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The effects of the tumor microenvironment the therapeutic efficacy of combining chemotherapy with checkpoint inhibitors in patients with lung cancer harboring rare -driver mutations remain unclear. We utilized single-cell RNA- and T-cell receptor (TCR) -sequencing to explore the immune and stromal cell profiles of 12 tumors and five tumor-adjacent tissues in seven patients with non-small cell lung cancer (NSCLCs) with rare -driver mutations treated with anti-PD-1 agents combined with chemotherapy. A class of highly expanded T -cells, known as GZMK + CD8+ effector memory T cells (GZMK + CD8+Tem), was enriched in both responsive tumors with and without rare driver mutations, suggesting similar anti-tumor immune mechanisms in both cohorts and that high levels of GZMK + CD8+Tem might be associated with effective responses to combination therapy. Non-responsive tumors exhibited a highly immunosuppressive M2-phenotype with enriched macrophages and monocytes. In non-major pathological response tumors, tumor cells interacted with alveolar and M0 macrophages via LAMC2-(ITGA6+ITGB1), possibly leading to M2 polarization. OAS1 was specifically expressed in CHIT1+ and FABP4+ macrophages and promoted macrophage polarization. These findings suggest that combination therapy reprogramed alveolar and M0-like macrophages to a pro-tumor phenotype, creating an immunosuppressive tumor microenvironment that resisted anti-PD1 therapy. In conclusion, GZMK + CD8+Tem is crucial for effective responses, whereas myeloid cells contribute to the immunosuppressive effects in anti-PD-1 therapies for NSCLCs with rare-driver mutations.
Collapse
Affiliation(s)
- Rui Fu
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | | | - Xuan Gao
- Geneplus-Beijing Institute, Beijing, China
| | - Fang Li
- Geneplus-Beijing Institute, Beijing, China
| | - Miao Cai
- Geneplus-Beijing Institute, Beijing, China
| | - Ben-Yuan Jiang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Shi Z, Shao K, Wang K, Xu M, Yu X, Xu C, Li Q, Song Z. Post-marketing safety of pralsetinib: a real-world disproportionality analysis based on the FDA adverse event reporting system database. Int J Clin Pharm 2025:10.1007/s11096-025-01917-z. [PMID: 40285824 DOI: 10.1007/s11096-025-01917-z] [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: 09/21/2024] [Accepted: 03/29/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Pralsetinib is a novel rearranged during transfection (RET) inhibitor that is approved for treating non-small cell lung cancer and thyroid cancer. Although clinical trials have established its efficacy, real-world data on its safety profile remain limited. AIM This study aimed to analyze pralsetinib-related adverse events (AEs) reported in the Food and Drug Administration adverse event reporting system (FAERS) database to identify potential safety signals. METHOD We conducted a retrospective pharmacovigilance analysis using FAERS database from Q3 2020 to Q2 2024. After deduplication, disproportionality analysis was performed using four algorithms: reporting odds ratio (ROR), proportional reporting ratio, Bayesian confidence propagation neural network, and empirical Bayes geometric mean. RESULTS A total of 1064 pralsetinib-related reports were identified, encompassing 3608 AEs. The most common AEs were hypertension (n = 80), asthenia (n = 79), anemia (n = 65), white blood cell count decreased (n = 63), and constipation (n = 58). We also detected new and unexpected AE signals, including blood calcitonin increased (ROR: 853.54), myocardial necrosis marker increased (ROR: 201.79), cystitis bacterial (ROR: 134.84), fungal foot infection (ROR: 51.83), pulmonary tuberculosis (ROR: 39.5), and myocardial injury (ROR: 30.36). Additionally, hypertension was more prevalent among female patients (Female/Male = 53/19, ROR: 1.8 [1.06-3.05]) and olderpatients (≥ 65/ < 65 = 34/18, ROR: 1.8 [1.01-3.2]). CONCLUSION Our study identified some known and new significant AE signals associated with pralsetinib, emphasizing the importance of continued pharmacovigilance. While the findings provide valuable insights for clinical practice, further validation through large-scale prospective studies is needed.
Collapse
Affiliation(s)
- Zheng Shi
- Department of Clinical Trial, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China
- Wenzhou Medical University, Wenzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Keda Shao
- Department of Clinical Trial, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China
- Wenzhou Medical University, Wenzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Ke Wang
- Department of Clinical Trial, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China
- Zhejiang Chinese Medical University, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Manyi Xu
- Department of Clinical Trial, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China
- Zhejiang Chinese Medical University, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xiayao Yu
- Department of Clinical Trial, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China
- Wenzhou Medical University, Wenzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Chunwei Xu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Qin Li
- Department of Clinical Trial, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Zhengbo Song
- Department of Clinical Trial, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China.
- Wenzhou Medical University, Wenzhou, China.
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
3
|
Sreenivasan S, Jiwani RA, White R, Bakalov V, Moll R, Liput J, Greenberg L. Advances in Targeted and Systemic Therapy for Salivary Gland Carcinomas: Current Options and Future Directions. Curr Oncol 2025; 32:232. [PMID: 40277788 PMCID: PMC12025620 DOI: 10.3390/curroncol32040232] [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: 03/10/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
Salivary gland carcinomas (SGCs) represent a rare and heterogeneous group of malignancies accounting for 3-6% of all head and neck cancers. While surgical resection and radiotherapy remain the standard for locoregional control, systemic treatment is indicated for recurrent or metastatic disease. Advances in molecular profiling have identified actionable targets such as NTRK gene fusions, HER2, immune checkpoint regulators, androgen receptors, and RET receptors. These have facilitated the development of targeted therapies, including TRK inhibitors, HER2-directed agents, and androgen receptor modulators, as well as emerging combinations of immunotherapy and chemotherapy. Despite these advancements, challenges such as resistance mechanisms and limited therapeutic efficacy persist. Overall response rates remain relatively low across most systemic therapies, reflecting a persistent unmet clinical need. This review discusses the current landscape of treatment options and explores promising clinical trials and future directions to enhance outcomes for patients with SGCs.
Collapse
Affiliation(s)
- Sushanth Sreenivasan
- Division of Internal Medicine, Allegheny Health Network, 320 East North Ave, Pittsburgh, PA 15212, USA
| | - Rahim A. Jiwani
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| | - Richard White
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| | - Veli Bakalov
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| | - Ryan Moll
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| | - Joseph Liput
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| | - Larisa Greenberg
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| |
Collapse
|
4
|
Liu SV, Nagasaka M, Atz J, Solca F, Müllauer L. Oncogenic gene fusions in cancer: from biology to therapy. Signal Transduct Target Ther 2025; 10:111. [PMID: 40223139 PMCID: PMC11994825 DOI: 10.1038/s41392-025-02161-7] [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: 01/29/2024] [Revised: 12/06/2024] [Accepted: 01/16/2025] [Indexed: 04/15/2025] Open
Abstract
Oncogenic gene fusions occur across a broad range of cancers and are a defining feature of some cancer types. Cancers driven by gene fusion products tend to respond well to targeted therapies, where available; thus, detection of potentially targetable oncogenic fusions is necessary to select optimal treatment. Detection methods include non-sequencing methods, such as fluorescence in situ hybridization and immunohistochemistry, and sequencing methods, such as DNA- and RNA-based next-generation sequencing (NGS). While NGS is an efficient way to analyze multiple genes of interest at once, economic and technical factors may preclude its use in routine care globally, despite several guideline recommendations. The aim of this review is to present a summary of oncogenic gene fusions, with a focus on fusions that affect tyrosine kinase signaling, and to highlight the importance of testing for oncogenic fusions. We present an overview of the identification of oncogenic gene fusions and therapies approved for the treatment of cancers harboring gene fusions, and summarize data regarding treating fusion-positive cancers with no current targeted therapies and clinical studies of fusion-positive cancers. Although treatment options may be limited for patients with rare alterations, healthcare professionals should identify patients most likely to benefit from oncogenic gene fusion testing and initiate the appropriate targeted therapy to achieve optimal treatment outcomes.
Collapse
Affiliation(s)
- Stephen V Liu
- Division of Hematology and Oncology, Georgetown University, Washington, DC, USA.
| | - Misako Nagasaka
- Division of Hematology Oncology, Department of Medicine, University of California Irvine School of Medicine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, Orange, CA, USA
| | - Judith Atz
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - Flavio Solca
- Boehringer Ingelheim RCV GmbH & Co.KG, Vienna, Austria
| | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
| |
Collapse
|
5
|
Zieliński P, Stępień M, Chowaniec H, Kalyta K, Czerniak J, Borowczyk M, Dwojak E, Mroczek M, Dworacki G, Ślubowska A, Markiewicz H, Ałtyn R, Dobosz P. Resistance in Lung Cancer Immunotherapy and How to Overcome It: Insights from the Genetics Perspective and Combination Therapies Approach. Cells 2025; 14:587. [PMID: 40277912 PMCID: PMC12026305 DOI: 10.3390/cells14080587] [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: 03/11/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
Lung cancer with the highest number of new cases diagnosed in Europe and in Poland, remains an example of malignancy with a very poor prognosis despite the recent progress in medicine. Different treatment strategies are now available for cancer therapy based on its type, molecular subtype and other factors including overall health, the stage of disease and cancer molecular profile. Immunotherapy is emerging as a potential addition to surgery, chemotherapy, radiotherapy or other targeted therapies, but also considered a mainstay therapy mode. This combination is an area of active investigation in order to enhance efficacy and overcome resistance. Due to the complexity and dynamic of cancer's ecosystem, novel therapeutic targets and strategies need continued research into the cellular and molecular mechanisms within the tumour microenvironment. From the genetic point of view, several signatures ranging from a few mutated genes to hundreds of them have been identified and associated with therapy resistance and metastatic potential. ML techniques and AI can enhance the predictive potential of genetic signatures and model the prognosis. Here, we present the overview of already existing treatment approaches, the current findings of key aspects of immunotherapy, such as immune checkpoint inhibitors (ICIs), existing molecular biomarkers like PD-L1 expression, tumour mutation burden, immunoscore, and neoantigens, as well as their roles as predictive markers for treatment response and resistance.
Collapse
Affiliation(s)
- Paweł Zieliński
- Chair of Pathomorphology and Clinical Immunology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (H.C.); (J.C.); (E.D.); (G.D.); (P.D.)
| | - Maria Stępień
- Université Paris-Saclay, UVSQ, INSERM, END-ICAP, 94805 Versailles, France;
- Doctoral School, Medical University of Lublin, 20-954 Lublin, Poland
| | - Hanna Chowaniec
- Chair of Pathomorphology and Clinical Immunology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (H.C.); (J.C.); (E.D.); (G.D.); (P.D.)
| | - Kateryna Kalyta
- Faculty of Biology, University of Basel, 4123 Basel, Switzerland;
| | - Joanna Czerniak
- Chair of Pathomorphology and Clinical Immunology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (H.C.); (J.C.); (E.D.); (G.D.); (P.D.)
| | - Martyna Borowczyk
- Department of Endocrinology, Internal Medicine and Metabolism, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Ewa Dwojak
- Chair of Pathomorphology and Clinical Immunology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (H.C.); (J.C.); (E.D.); (G.D.); (P.D.)
- Department of Pathomorphology, University Clinical Hospital, 61-701 Poznan, Poland
| | - Magdalena Mroczek
- Department of Neurology, University Hospital Basel, 4123 Basel, Switzerland;
| | - Grzegorz Dworacki
- Chair of Pathomorphology and Clinical Immunology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (H.C.); (J.C.); (E.D.); (G.D.); (P.D.)
| | - Antonina Ślubowska
- Department of Biostatistics and Research Methodology, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University of Warsaw, 02-004 Warsaw, Poland;
| | - Hanna Markiewicz
- Department of Histology and Embryology, Faculty of Medicine, Medical University of Warsaw, 02-004 Warsaw, Poland
- Department of Methodology, Faculty of Medicine, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Rafał Ałtyn
- IT Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Paula Dobosz
- Chair of Pathomorphology and Clinical Immunology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (H.C.); (J.C.); (E.D.); (G.D.); (P.D.)
| |
Collapse
|
6
|
Tu DH, Qu R, Wen F, Zhou Q, Liu Q, Huang L, Chen T. Successful conversion surgery following tislelizumab with chemotherapy in a patient with stage IIIC lung adenocarcinoma harboring RET fusions: A case report and review of the literature. Exp Ther Med 2025; 29:70. [PMID: 39991722 PMCID: PMC11843209 DOI: 10.3892/etm.2025.12820] [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: 08/15/2024] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have emerged as a beacon of hope for most patients with stage III non-small cell lung cancer (NSCLC) who are no longer surgical candidates. However, the literature on the use of immunotherapy in patients with NSCLC with rearranged during transfection (RET) gene fusions is scant. The present study reports the case of a 61-year-old female patient, diagnosed with stage IIIC lung adenocarcinoma, exhibiting two RET gene fusions and high programmed death-ligand 1 expression. Following four treatment cycles of tislelizumab in combination with pemetrexed and cisplatin, the patient was successfully downstaged, enabling radical surgery. The post-operative pathology analysis indicated a major pathologic response. This case study contributes to the growing body of evidence supporting the use of ICIs in treating locally advanced NSCLC with RET gene fusions.
Collapse
Affiliation(s)
- De-Hao Tu
- Department of Thoracic Surgery, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Rirong Qu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Fang Wen
- Department of Oncology, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Qiang Zhou
- Department of Oncology, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Qianyun Liu
- Department of Medical Imaging, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Lingmei Huang
- Department of Pulmonary and Critical Care Medicine, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Tao Chen
- Department of Thoracic Surgery, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| |
Collapse
|
7
|
Zhu T, Zhong J, Huang Y. Cancer Drugs Approved Based on Surrogate Endpoint: A Retrospective Observational Study in the United States and China. Cancer Med 2025; 14:e70864. [PMID: 40230311 PMCID: PMC11997453 DOI: 10.1002/cam4.70864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Hundreds of cancer drugs are approved globally based on surrogate endpoint response rate (RR). However, the characteristics of RR-based approvals remain unknown. METHODS In this retrospective study, all cancer drug-indication pairs approved based on RR in the United States and China up to December 2023 were analyzed. RESULTS A total of 249 RR-supported drug-indication pairs were identified in the United States and 98 in China. In the United States, 98 of the 249 (39.4%) indications were granted regular approval (RA), whereas in China, only 21 of 98 (21.4%) approvals followed this regulatory pathway, with the remainder receiving accelerated approval (AA). The conversion rate from AA to RA was significantly lower in China compared to the United States (13.3% vs. 28.1%, p < 0.001). The proportion of AA withdrawals was significantly lower in China compared to the United States (1.0% vs. 10.4%, p < 0.001). Among all indications, the median RR in China was 60.9% (IQR, 35.8%-75.0%), which was significantly higher than the 45.0% (IQR, 29.0%-61.0%) in the United States (p < 0.001). In China, 18 of the 98 (18.4%) had an RR less than 30%. In contrast, in the United States, 26.9% of the 249 had an RR less than 30%. CONCLUSIONS Compared to the United States, RR-supported approvals in China are characterized by higher RR values and a stricter RA pathway. Regulatory authorities in both countries may need to consider both the quantity and quality during cancer drug development based on surrogate endpoints.
Collapse
Affiliation(s)
- Ting Zhu
- School of General Practice and Continuing EducationCapital Medical UniversityBeijingChina
| | - Jinjia Zhong
- School of General Practice and Continuing EducationCapital Medical UniversityBeijingChina
| | - Yafang Huang
- School of General Practice and Continuing EducationCapital Medical UniversityBeijingChina
| |
Collapse
|
8
|
Chung C, Umoru G. Prognostic and predictive biomarkers with therapeutic targets in nonsmall-cell lung cancer: A 2023 update on current development, evidence, and recommendation. J Oncol Pharm Pract 2025; 31:438-461. [PMID: 38576390 DOI: 10.1177/10781552241242684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
BackgroundSince the publication of the original work in 2014, significant progress has been made in the characterization of genomic alterations that drive oncogenic addiction of nonsmall cell lung cancer (NSCLC) and how the immune system can leverage non-oncogenic pathways to modulate therapeutic outcomes. This update evaluates and validates the recent and emerging data for prognostic and predictive biomarkers with therapeutic targets in NSCLC.Data sourcesWe performed a literature search from January 2015 to October 2023 using the keywords non-small cell lung cancer, clinical practice guidelines, gene mutations, genomic assay, immune cancer therapy, circulating tumor DNA, predictive and prognostic biomarkers, and targeted therapies.Study selection and data extractionWe identified, reviewed, and evaluated relevant clinical trials, meta-analyses, seminal articles, and published clinical practice guidelines in the English language.Data synthesisRegulatory-approved targeted therapies include those somatic gene alterations of EGFR ("classic" mutations, exon 20 insertion, and rare EGFR mutations), ALK, ROS1, BRAF V600, RET, MET, NTRK, HER2, and KRAS G12C. Data for immunotherapy and circulating tumor DNA in next-generation sequencing are considered emerging, whereas the predictive role for PIK3CA gene mutation is insufficient.ConclusionsAdvances in sequencing and other genomic technologies have led to identifying novel oncogenic drivers, novel resistance mechanisms, and co-occurring mutations that characterize NSCLC, creating further therapeutic opportunities. The benefits associated with immunotherapy in the perioperative setting hold initial promise, with their long-term results awaiting.
Collapse
Affiliation(s)
- Clement Chung
- Department of Pharmacy, Houston Methodist West Hospital, Houston, TX, USA
| | - Godsfavour Umoru
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
9
|
Sullo FG, Garinet S, Blons H, Taieb J, Laurent-Puig P, Gallois C. Molecular features and clinical actionability of gene fusions in colorectal cancer. Crit Rev Oncol Hematol 2025; 208:104656. [PMID: 39922396 DOI: 10.1016/j.critrevonc.2025.104656] [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: 09/24/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer death and accounts for 10 % of cancer diagnoses worldwide. Despite the advancements achieved over the latest decades, CRC treatments are still based on conventional chemotherapy whose efficacy is limited by acquired resistance and unfavorable toxicity profile, making the search for novel actionable targets a priority. In this context, gene fusions are emerging as promising -albeit very rare - new markers because of their recurrence across different tumor types and their potential actionability. The aim of this review is to investigate the role of gene fusions in CRC by focusing on pathogenesis, screening strategies as well as their clinical implications.
Collapse
Affiliation(s)
- Francesco Giulio Sullo
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Institut du Cancer Paris CARPEM, Paris, France; Institut du Cancer Paris CARPEM, AP-HP.Centre, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Simon Garinet
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Institut du Cancer Paris CARPEM, Paris, France; APHP.Centre, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France
| | - Hélène Blons
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Institut du Cancer Paris CARPEM, Paris, France; APHP.Centre, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France
| | - Julien Taieb
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Institut du Cancer Paris CARPEM, Paris, France; Institut du Cancer Paris CARPEM, AP-HP.Centre, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Institut du Cancer Paris CARPEM, Paris, France; APHP.Centre, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France
| | - Claire Gallois
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Institut du Cancer Paris CARPEM, Paris, France; Institut du Cancer Paris CARPEM, AP-HP.Centre, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France.
| |
Collapse
|
10
|
Galeș LN, Păun MA, Butnariu I, Simion L, Manolescu LSC, Trifănescu OG, Anghel RM. Next-Generation Sequencing in Oncology-A Guiding Compass for Targeted Therapy and Emerging Applications. Int J Mol Sci 2025; 26:3123. [PMID: 40243903 PMCID: PMC11988731 DOI: 10.3390/ijms26073123] [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: 02/14/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Multigene sequencing technologies provide a foundation for targeted therapy and precision oncology by identifying actionable alterations and enabling the development of treatments that substantially improve clinical outcomes. This review emphasizes the importance of having a molecular compass guiding treatment decision-making through the multitude of alterations and genetic mutations, showcasing why NGS plays a pivotal role in modern oncology.
Collapse
Affiliation(s)
- Laurenția Nicoleta Galeș
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- Department of Medical Oncology II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Mihai-Andrei Păun
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
| | - Ioana Butnariu
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Laurentiu Simion
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Loredana Sabina Cornelia Manolescu
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- Clinical Laboratory of Medical Microbiology, “Marius Nasta” Institute of Pneumology, 050159 Bucharest, Romania
- Department of Microbiology, Parasitology and Virology, Faculty of Midwives and Nursing, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Oana Gabriela Trifănescu
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- Department of Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Rodica Maricela Anghel
- “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.)
- Department of Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| |
Collapse
|
11
|
Katayama Y, Yamada T, Tanimura K, Kawachi H, Ishida M, Matsui Y, Hirai S, Nakamura R, Morimoto K, Furuya N, Arai S, Goto Y, Sakata Y, Nishino K, Tsuchiya M, Tamiya A, Saito G, Muto S, Takeda T, Date K, Fujisaka Y, Watanabe S, Fujimoto D, Uehara H, Horinaka M, Sakai T, Yano S, Tokuda S, Takayama K. YAP Regulates HER3 Signaling-Driven Adaptive Resistance to RET Inhibitors in RET-Aberrant Cancers. Clin Cancer Res 2025; 31:1127-1141. [PMID: 39495173 DOI: 10.1158/1078-0432.ccr-24-1762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/06/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
PURPOSE Rearranged during transfection (RET) aberrations represent a targetable oncogene in several tumor types, with RET inhibitors displaying marked efficacy. However, some patients with RET-aberrant cancer are insensitive to RET tyrosine kinase inhibitors (TKI). Recently, drug-tolerant mechanisms have attracted attention as targets for initial therapies to overcome drug resistance. The underlying mechanisms of drug-tolerant cell emergence treated with RET-TKIs derived from RET-aberrant cancer cells remain unknown. This study investigated the role of YAP-mediated HER3 signaling in the underlying mechanisms of adaptive resistance to RET-TKIs in RET-aberrant cancer cells. EXPERIMENTAL DESIGN Four RET-aberrant cancer cell lines were used to assess sensitivity to the RET-TKIs selpercatinib and pralsetinib and to elucidate the molecular mechanisms underlying adaptive resistance using RNA sequencing, phospho-receptor tyrosine kinase antibody arrays, chromatin immunoprecipitation assay, and luciferase reporter assays. Clinical specimens from patients with RET fusion-positive lung cancer were analyzed for pretreatment YAP expression and correlated with treatment outcomes. RESULTS In high YAP-expressing RET-aberrant cancer cells, YAP-mediated HER3 signaling activation maintained cell survival and induced the emergence of cells tolerant to the RET-TKIs selpercatinib and pralsetinib. The pan-ErBB inhibitor afatinib and YAP/tea domain inhibitors verteporfin and K-975 sensitized YAP-expressing RET-aberrant cancer cells to the RET-TKIs selpercatinib and pralsetinib. Pretreatment YAP expression in clinical specimens obtained from patients with RET fusion-positive lung cancer was associated with poor RET-TKI treatment outcomes. CONCLUSIONS The YAP-HER3 axis is crucial for the survival and adaptive resistance of high YAP-expressing RET-aberrant cancer cells treated with RET-TKIs. Combining YAP/HER3 inhibition with RET-TKIs represents a highly potent strategy for initial treatment. See related commentary by Ortiz-Cuaran and Leonce, p. 958.
Collapse
Affiliation(s)
- Yuki Katayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tadaaki Yamada
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keiko Tanimura
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hayato Kawachi
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masaki Ishida
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yohei Matsui
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Soichi Hirai
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryota Nakamura
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenji Morimoto
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Naoki Furuya
- Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of Medicine, Kawasaki, Japan
| | - Sachiko Arai
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Yasuhiro Goto
- Department of Respiratory Medicine and Allergies, Fujita Health University, Toyoake, Japan
| | - Yoshihiko Sakata
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Kazumi Nishino
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Michiko Tsuchiya
- Department of Respiratory Medicine, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - Akihiro Tamiya
- Department of Internal Medicine, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai, Japan
| | - Go Saito
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Muto
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Takayuki Takeda
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Koji Date
- Department of Pulmonary Medicine, Kyoto Chubu Medical Center, Nantan, Japan
| | - Yasuhito Fujisaka
- Department of Respiratory Medicine and Thoracic Oncology, Clinical Research Center, Osaka Medical and Pharmaceutical University Hospital, Takatsuki, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Daichi Fujimoto
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Japan
| | - Hisanori Uehara
- Division of Pathology, Tokushima University Hospital, Tokushima, Japan
| | - Mano Horinaka
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiyuki Sakai
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Seiji Yano
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
- Department of Respiratory Medicine, Kanazawa Graduate School of Medical Sciences, Kanazawa, Japan
| | - Shinsaku Tokuda
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
12
|
Xu M, Wu K, He R, He J, Yang G, Ma H, Peng L, Zhang S, Tan L, Zhang Z, Cai Q. Design, synthesis and evaluation of (E)-1-(4-(2-(1H-pyrazol-5-yl)vinyl)phenyl) derivatives as next generation selective RET inhibitors overcoming RET solvent front mutations (G810C/R). Eur J Med Chem 2025; 286:117294. [PMID: 39879936 DOI: 10.1016/j.ejmech.2025.117294] [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: 12/01/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/31/2025]
Abstract
RET is a well-recognized drug target for cancer treatment. Despite the promising efficacy of selective second-generation RET inhibitors Selpercatinib and Pralsetinib, the clinical benefits have been compromised due to the quickly developed resistance to these drugs. RET G810 mutations at the solvent front site have been identified as the major on-target mutations contributing to resistance against Selpercatinib and Pralsetinib. Therefore, there is an urgent need for the development of next-generation RET inhibitors to overcome acquired solvent-front resistance mutations. In this study, a series of (E)-1-(4-(2-(1H-pyrazol-5-yl)vinyl)phenyl) derivatives have been identified as selective next-generation RET inhibitors. The representative compound, CQ1373 exhibits potent cellular potency with IC50 values of 13.0, 25.7 and 28.4 nM against BaF3 cells expressing CCDC6-RET, CCDC6-RET-G810C and CCDC6-RET-G810R, respectively. A comprehensive selectivity profile across 89 kinases reveals that CQ1373 demonstrates good selectivity toward wild-type RET and solvent front mutants G810C/R with IC50 values of 4.2, 7.1 and 32.4 nM, respectively. Furthermore, western blot analysis reveals that CQ1373 effectively inhibits RET phosphorylation and downstream signaling through SHC. It also induces apoptosis and cell cycle arrest in a dose-dependent manner in BaF3 cells harboring CCDC6-RET, CCDC6-RET-G810C and CCDC6-RET-G810R fusions. More significantly, CQ1373 exhibits promising in vivo anti-tumor efficacy in a CCDC6-RET-G810R mice xenograft model, highlighting its potentials for RET-driven cancers treatment.
Collapse
Affiliation(s)
- Mingjin Xu
- College of Chemistry and Materials Science, Zhejiang Normal University, No. 688 Yingbin Road, Jinhua, Zhejiang Province, 321004, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Kaifu Wu
- College of Chemistry and Materials Science, Zhejiang Normal University, No. 688 Yingbin Road, Jinhua, Zhejiang Province, 321004, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Rui He
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, School of Pharmacy, Jinan University, Guangzhou, 510632, China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Jiahuan He
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, School of Pharmacy, Jinan University, Guangzhou, 510632, China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Gangpeng Yang
- College of Chemistry and Materials Science, Zhejiang Normal University, No. 688 Yingbin Road, Jinhua, Zhejiang Province, 321004, China
| | - Haowen Ma
- College of Chemistry and Materials Science, Zhejiang Normal University, No. 688 Yingbin Road, Jinhua, Zhejiang Province, 321004, China
| | - Lijie Peng
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510632, China
| | - Li Tan
- College of Chemistry and Materials Science, Zhejiang Normal University, No. 688 Yingbin Road, Jinhua, Zhejiang Province, 321004, China.
| | - Zhang Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, School of Pharmacy, Jinan University, Guangzhou, 510632, China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou, 510632, China; Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510632, China.
| | - Qian Cai
- College of Chemistry and Materials Science, Zhejiang Normal University, No. 688 Yingbin Road, Jinhua, Zhejiang Province, 321004, China.
| |
Collapse
|
13
|
Bouchard N, Daaboul N. Lung Cancer: Targeted Therapy in 2025. Curr Oncol 2025; 32:146. [PMID: 40136350 PMCID: PMC11941068 DOI: 10.3390/curroncol32030146] [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: 01/31/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
Lung cancer treatment has changed in the last twenty years since the discovery of EGFR mutations. In this article, we will review the current state of the art for non-small cell lung cancer (NSCLC) actionable genomic alterations (AGA). AGAs are mostly found in lung adenocarcinomas, a subtype of non-small cell lung cancers. We will focus on the current treatment for EGFR mutations, ALK fusions, ROS1 fusions, BRAF V600E mutations, MET exon 14-skipping mutations, RET fusions, KRAS G12C mutations, ERBB2 mutations (also called HER2 mutations), and NTRK fusions. We will also touch on the key toxicities associated with these medications. Treatments are mostly available for the metastatic stage, but we will also discuss adjuvant therapy for EGFR mutations and ALK fusions, as well as stage III post-chemoradiotherapy treatment for EGFR lung cancer.
Collapse
Affiliation(s)
- Nicole Bouchard
- Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Nathalie Daaboul
- Centre Intégré de Cancérologie de la Montérégie, Université de Sherbrooke, Longueuil, QC J4V 2H1, Canada
| |
Collapse
|
14
|
Gritsch D, Brastianos PK. Molecular evolution of central nervous system metastasis and therapeutic implications. Trends Mol Med 2025; 31:240-251. [PMID: 39424530 PMCID: PMC11908961 DOI: 10.1016/j.molmed.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024]
Abstract
The increasing prevalence and poor prognosis of central nervous system (CNS) metastases pose a significant challenge in oncology, necessitating improved therapeutic strategies. Recent research has shed light on the complex genomic landscape of brain metastases, identifying unique and potentially actionable genetic alterations. These insights offer new avenues for targeted therapy, highlighting the potential of precision medicine approaches in treating CNS metastases. However, translating these discoveries into clinical practice requires overcoming challenges such as availability of tissue for characterization, access to molecular testing, drug delivery across the blood-brain barrier (BBB) and addressing intra- and intertumoral genetic heterogeneity. This review explores novel insights into the evolution of CNS metastases, the molecular mechanisms underlying their development, and implications for therapeutic interventions.
Collapse
Affiliation(s)
- David Gritsch
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Priscilla K Brastianos
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA.
| |
Collapse
|
15
|
Tang X, Hu J, Guo R, Wang Y, Yao Y, Zeng L, Wang H, Shen H, Chen J, Huang W, Liu L, Su M, Yu Z, Mao W, Wang J, Tao H, Dong G, Cai C, Xie Y, Qu T, Zhang Y, Shen W, Cao J, Cai X, Kong W, Li X, Liang F. RET-AREAL: A multi-center, real-world data analysis on the efficacy of pralsetinib in acquired RET fusion after resistance to EGFR/ALK-TKIs. Cancer Lett 2025; 612:217455. [PMID: 39800214 DOI: 10.1016/j.canlet.2025.217455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/31/2024] [Accepted: 01/10/2025] [Indexed: 01/15/2025]
Abstract
Pralsetinib demonstrated impressive improvement of survival in non-small cell lung cancer (NSCLC) patients harbored de novo RET fusion. However, the efficacy in patients with acquired RET fusion after resistance to EGFR/ALK-TKIs has only been reported on a case-by-case basis, and the strategy for overcoming the acquired RET fusion has not been fully investigated. This multicenter, real-world analysis enrolled 32 patients with unresectable NSCLC harbored acquired RET fusion after resistance to EGFR/ALK-TKIs in 23 centers across China from July 1st, 2018 to Nov 23rd, 2022. Epidemiological, clinical, genetic, and treatment data were collected. The primary outcome was time to treatment failure (TTF). Secondary outcomes were overall survival (OS), objective response rate (ORR), disease control rate (DCR) and toxicity. In real-world context, patients underwent pralsetinib-based treatment had a higher proportion of central nervous system metastasis. EGFR 19del was the predominant mutation type (62.5 %) prior to acquired RET fusion. CCDC6 was the commonest RET fusion partner (40.6 %). "Clonal RET fusion" (c-RET) and "subclonal RET fusion" (s-RET) were defined according to the RET fusion allele frequency. Patients with c-RET had higher proportions of undetected EGFR mutation and KIF5B-RET fusion. First-line pralsetinib-based therapy had notably superior median TTF when compared to their counterparts (8.03 versus 4.30 months, P = 0.016). Notably, patients with c-RET had a better prognosis than those with s-RET (median TTF: NR versus 5.67 months, P = 0.037, median OS: NR versus 9.83 months, P = 0.047). In conclusion, pralsetinib-based therapy may be a potential strategy to overcome acquired RET fusion after resistance to EGFR/ALK-TKIs.
Collapse
Affiliation(s)
- Xinjun Tang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Hu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pulmonary and Critical Care Medicine, Shanghai Geriatrics Medical Center, Shanghai, China.
| | - Renhua Guo
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yina Wang
- Department of Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Yao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Zeng
- Department of Medical Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Haitao Wang
- Department of Oncology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haibo Shen
- Department of Thoracic Oncology, Ningbo No.2 Hospital, Ningbo, China
| | - Jun Chen
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Weina Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Lihua Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ming Su
- Department of Lung Disease, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Zhuang Yu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Weidong Mao
- Department of Oncology, Jiangyin People's Hospital, Wuxi, China
| | - Jinliang Wang
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, China
| | - Haitao Tao
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, China
| | - Guilan Dong
- Department of Oncology, Tangshan People's Hospital, Tangshan, China
| | - Chang Cai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yupeng Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Qu
- Department of Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Zhang
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Wei Shen
- Department of Pulmonary and Critical Care Medicine, The Third People's Hospital of Cixi, Ningbo, China
| | - Jing Cao
- Department of Pulmonary and Critical Care Medicine, Yiyang Central Hospital, Yiyang, China
| | - Xin Cai
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wencui Kong
- Department of Pulmonary and Critical Care Medicine, The 900th Hospital of the Joint Logistic Support Force, People's Liberation Army of China, Fuzhou, China
| | - Xiaoxue Li
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fei Liang
- Department of Biostatistics, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Ruan DY, Huang WW, Li Y, Zhao Y, Shi Y, Jia Y, Cang S, Zhang W, Shi J, Chen J, Lin J, Liu Y, Xu J, Ouyang W, Fang J, Zhuang W, Liu C, Bu Q, Li M, Meng X, Sun M, Yang N, Dong X, Pan Y, Li X, Qu X, Zhang T, Yuan X, Hu S, Guo W, Li Y, Li S, Liu D, Song F, Tan L, Yu Y, Yu X, Zang A, Sun C, Zhang Q, Zou K, Dan M, Xu RH, Zhao H. Safety, pharmacokinetics and efficacy of HA121-28 in patients with advanced solid tumors and RET fusion-positive non-small-cell lung cancer: a multicenter, open-label, single-arm phase 1/2 trial. Signal Transduct Target Ther 2025; 10:62. [PMID: 40016191 PMCID: PMC11868595 DOI: 10.1038/s41392-025-02155-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/20/2024] [Accepted: 01/22/2025] [Indexed: 03/01/2025] Open
Abstract
HA121-28, a promising multikinase inhibitor, mainly targets rearranged during transfection (RET) fusions and selectively targets vascular endothelial growth factor receptor-2, endothelial growth factor receptor, and fibroblast growth factor receptor 1-3. The safety, pharmacokinetics, and efficacy of HA121-28 were assessed in advanced solid tumors (phase 1, ClinicalTrials.gov NCT03994484) and advanced RET fusion-positive non-small-cell lung cancer (RET-TKI naive NSCLC, phase 2, ClinicalTrials.gov NCT05117658). HA121-28 was administered orally in doses range from 25 to 800 mg under the 21-day on/7-day off scheme for a 28-day cycle in phase 1 trial. The recommended dose identified in phase 1 (450 mg) was administered for patients during phase 2. The primary endpoints were the maximum tolerated dose (MTD) in phase 1 and the objective response rate (ORR) in phase 2. 162 patients were enrolled in phase 1 and 48 in phase 2. A total of 600 mg once daily was set as MTD. Across 100-800 mg, the exposure of HA121-28 increased in a dose-dependent manner. Consistent between both trials, diarrhea, rash, and prolonged QTc interval, were the most reported treatment-emergent adverse events. 40.0% (phase 1) and 62.5% (phase 2) patients experienced grade ≥3 treatment-related adverse events, respectively. The overall ORR was 26.8% and the median progression-free survival (PFS) was 5.5 months among 97 NSCLC patients with advanced RET fusion receiving a dose at ≥450 mg once daily. HA121-28 showed encouraging efficacy in advanced RET fusion NSCLC and its toxicity was tolerable in most patients. Nevertheless, cardiotoxicity is a notable concern that warrants careful attention.
Collapse
Affiliation(s)
- Dan-Yun Ruan
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wen-Wen Huang
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yongsheng Li
- Department of Phase 1 Ward, Chongqing University Cancer Hospital, Chongqing Cancer Hospital, Chongqing, People's Republic of China
| | - Yanqiu Zhao
- Department of Respiratory Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yehui Shi
- Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China
| | - Yuming Jia
- Department of Oncology, The second people's hospital of Yibin, Yibin, Sichuan, People's Republic of China
| | - Shundong Cang
- Department of Medical Oncology, Phase 1 Clinical Research Unit, Department of Medical Oncology, Henan Provincial People's Hospital, Zhengzhou, Hannan, People's Republic of China
| | - Wei Zhang
- Department of Medical Oncology, Phase 1 Clinical Research Unit, Department of Medical Oncology, Henan Provincial People's Hospital, Zhengzhou, Hannan, People's Republic of China
| | - Jianhua Shi
- Department of the Second General Medicine, Linyi Cancer Hospital, Linyi, Shandong, People's Republic of China
| | - Jun Chen
- Department of Pulmonary Oncology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Jie Lin
- Department of Oncology, The Second Affiliated hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jianming Xu
- Department of Medical Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Weiwei Ouyang
- The Phase1 Clinical Center, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Jian Fang
- Department of the Second Thoracic Oncology, Beijing Cancer Hospital, Beijing, People's Republic of China
| | - Wu Zhuang
- Department of Respiratory Oncology, Fujian Cancer Hospital, Fuzhou, Fujian, People's Republic of China
| | - Caigang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Qing Bu
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, People's Republic of China
| | - Xiangjiao Meng
- Department of the Fourth Thoracic Radiotherapy Ward, Shandong Cancer Hospital & Institute, Jinan, Shandong, People's Republic of China
| | - Meili Sun
- Department of Oncology, General Hospital Affiliated Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Nong Yang
- Department of Pulmonary and Gastrointestinal Medicine, Hunan Cancer Hospital, Changsha, Hunan, People's Republic of China
| | - Xiaorong Dong
- Department of Cancer Center, Wuhan Union Hospital of China, Wuhan, Hubei, People's Republic of China
| | - Yueyin Pan
- Department of Oncology Chemotherapy, The First Affiliated Hospital of USTC, Hefei, Anhui, People's Republic of China
| | - Xingya Li
- Department of the Second Oncology Ward, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Tongmei Zhang
- General Department, Beijing Chest Hospital, Beijing, People's Republic of China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College of HUST, Wuhan, Hubei, People's Republic of China
| | - Sheng Hu
- Department of Oncology, Hubei Cancer Hospital, Wuhan, Hubei, People's Republic of China
| | - Wei Guo
- Respiratory Department, Shanxi Cancer Hospital, Taiyuan, Shanxi, People's Republic of China
| | - Yalun Li
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shengqing Li
- Respiratory Department, Huashan Hospital Fudan University, Shanghai, People's Republic of China
| | - Dongying Liu
- Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, People's Republic of China
| | - Feixue Song
- Department of Medical Oncology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Liping Tan
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital & Guangxi Cancer Institute, Nanning, Guangxi, People's Republic of China
| | - Yan Yu
- Department of the Third Respiratory Medicine, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| | - Xinmin Yu
- Department of Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Aimin Zang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei, People's Republic of China
| | - Chang Sun
- CSPC ZhongQi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang, Hebei, People's Republic of China
| | - Qian Zhang
- CSPC ZhongQi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang, Hebei, People's Republic of China
| | - Kai Zou
- CSPC ZhongQi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang, Hebei, People's Republic of China
| | - Mo Dan
- CSPC ZhongQi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang, Hebei, People's Republic of China
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, People's Republic of China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, People's Republic of China.
| | - Hongyun Zhao
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
17
|
Zhao Z, Pang L, Liu S, Liu J. Re-challenging pralsetinib following recovery from pneumocystis jirovecii pneumonia in a lung cancer patient: a Case Report. Front Pharmacol 2025; 15:1443609. [PMID: 40083593 PMCID: PMC11904333 DOI: 10.3389/fphar.2024.1443609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/26/2024] [Indexed: 03/16/2025] Open
Abstract
Background Pneumocystis jirovecii pneumonia (PJP), an opportunistic infection, is commonly observed in immunocompromised individuals, particularly those with cancer, and is known for its significant morbidity and mortality rates. Pralsetinib is a highly specific inhibitor that targets advanced or metastatic non-small cell lung cancer (NSCLC) characterized by RET-fusion positivity. The incidence of PJP infection in patients receiving pralsetinib was found to be infrequent. However, there is currently a lack of consensus regarding the rechallenge of pralsetinib in patients who have fully recovered from PJP. Case presentation In this case study, a 60-year-old patient diagnosed with stage IV lung adenocarcinoma and carrying a KIF5B-RET fusion gene underwent pralsetinib treatment as the fourth-line therapy. Subsequently, the patient developed a fever and dyspnea 2.5 months later. However, the patient did not exhibit a positive response to the empirical antibiotic therapy administered. The computed tomography findings indicated widespread ground-glass opacities with numerous cystic lesions in both lungs, along with patchy consolidations in the lower right lung. The diagnosis of PJP was conclusively confirmed through bronchoalveolar lavage. The patient's condition was effectively treated with a combination of oral trimethoprim/sulfamethoxazole and intravenous caspofungin along with clindamycin. The patient fully recovered from PJP. Subsequently, he underwent a rechallenge with pralsetinib, and as of the latest follow-up, no evidence of progressive disease has been observed. Conclusion This case report emphasizes the significance for physicians to be cognizant of the potential hazard of PJP development in cancer patients undergoing pralsetinib treatment, particularly in those who are unresponsive to empirical antibiotic therapy. Prompt identification and timely intervention are essential to achieve better outcomes in patients with pralsetinib-induced PJP. Furthermore, it highlights the scenario where patients who have fully recovered from moderate-to-severe pralsetinib-induced PJP may undergo pralsetinib re-administration without requiring alternative treatment options.
Collapse
Affiliation(s)
- Zhe Zhao
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, Shandong, China
| | - Longbin Pang
- Pulmonary and Critical Care Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Surui Liu
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jie Liu
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, Shandong, China
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
18
|
Chen X, Wang G, Zhang J, Bao W, Cai J, Guo J, Lv T, Ye M. Pathologic complete response to pralsetinib in stage IV RET-positive non-small cell lung cancer: A case report. Respir Med Case Rep 2025; 54:102186. [PMID: 40123940 PMCID: PMC11928833 DOI: 10.1016/j.rmcr.2025.102186] [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: 11/02/2024] [Revised: 01/31/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Neoadjuvant therapy with tyrosine kinase inhibitors has been proposed as a feasible approach for downstaging potential resectable non-small cell lung cancer (NSCLC). Pralsetinib is a paradigm of precision medicine for cancers driven by mutant RET (rearranged during transfection). In this case, we reported dramatic response to Pralsetinib in a stage IV NSCLC patient with RET rearrangement. Strikingly, treatment with 10 months of Pralsetinib impended downstaging of the N2 lymph node and metastatic pleural disease. Histological examination of the surgically resected specimen indicated a pathologic complete response (pCR). The patient was recommended to continue Pralsetinib as an adjuvant therapy. This case highlighted potential application of Pralsetinib in locally advanced RET-positive NSCLC to prime surgical resection. Postoperative Pralsetinib adjuvant therapy should also be considered.
Collapse
Affiliation(s)
- Xinxin Chen
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Guoxin Wang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Jianfeng Zhang
- Department of Thoracic Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Wei Bao
- Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Jun Cai
- Department of Radiology and Medical Imaging Center, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Jing Guo
- Department of Nuclear Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Tangfeng Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| |
Collapse
|
19
|
Lim JU, Jung J, Kim YW, Kim CY, Lee SH, Park DW, Choi SI, Ji W, Yeo CD, Lee SH. Targeting the Tumor Microenvironment in EGFR-Mutant Lung Cancer: Opportunities and Challenges. Biomedicines 2025; 13:470. [PMID: 40002883 PMCID: PMC11852785 DOI: 10.3390/biomedicines13020470] [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: 01/10/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have transformed the treatment of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer. However, treatment resistance remains a major challenge in clinical practice. The tumor microenvironment (TME) is a complex system composed of tumor cells, immune and non-immune cells, and non-cellular components. Evidence indicates that dynamic changes in TME during TKI treatment are associated with the development of resistance. Research has focused on identifying how each component of the TME interacts with tumors and TKIs to understand therapeutic targets that could address TKI resistance. In this review, we describe how TME components, such as immune cells, fibroblasts, blood vessels, immune checkpoint proteins, and cytokines, interact with EGFR-mutant tumors and how they can promote resistance to TKIs. Furthermore, we discuss potential strategies targeting TME as a novel therapeutic approach.
Collapse
Affiliation(s)
- Jeong Uk Lim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yeon Wook Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Chi Young Kim
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sang Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Institute of Chest Diseases, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dong Won Park
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, Republic of Korea;
| | - Sue In Choi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Wonjun Ji
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 44610, Republic of Korea
| | - Chang Dong Yeo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03083, Republic of Korea
| | - Seung Hyeun Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
20
|
Mullally WJ, O'Leary CG, O'Byrne KJ. Rearranged during transfection (RET) lung cancer - Update on targeted therapies. Lung Cancer 2025; 200:108083. [PMID: 39827484 DOI: 10.1016/j.lungcan.2025.108083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/18/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
The enhanced comprehension of the molecular pathways underpinning oncogenesis in non-small cell lung cancer (NSCLC) has led to the advancement of personalized treatment for individuals with actionable mutations using targeted therapies. The rearranged during transfection (RET) proto-oncogene, is critical in the embryonic development of various tissues, including renal, neural, and neuroendocrine tissue. RET fusions have been observed in approximately 1-2% of NSCLC cases. Targeted therapies for NSCLC with RET alterations have progressed significantly over the past decade. While multikinase inhibitors (MKIs) faced limitations in efficacy and tolerability, the introduction of selective RET inhibitors (SRIs) such as selpercatininb and pralsetinib has transformed patient outcomes, resulting in deep and durable responses. Ongoing clinical trials are exploring their potential benefits in the neoadjuvant and adjuvant setting. Early phase clinical trials endeavor to demonstrate next-generation selective RET inhibitors can effectively overcome SRI resistance mechanisms, offer improved safety profiles, and enhance patient outcomes.
Collapse
Affiliation(s)
- W J Mullally
- Department of Medical Oncology, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia.
| | - C G O'Leary
- Department of Medical Oncology, Mater Misericordiae Hospital, South Brisbane, Queensland 4101, Australia
| | - K J O'Byrne
- Department of Medical Oncology, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia; The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia.
| |
Collapse
|
21
|
Gupta B, Borghaei L, Liu SV. NRG1 Fusions: The New Kid on the Block. Curr Oncol Rep 2025; 27:190-194. [PMID: 39888568 DOI: 10.1007/s11912-025-01640-y] [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] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
PURPOSE OF REVIEW Neuregulin 1 (NRG1) fusions are rare but actionable oncogenic drivers that occur in a variety of tumor types, including non-small cell lung cancer (NSCLC). These fusions lead to pathophysiologic activation of HER signaling pathways, promoting tumor growth, invasion, and metastasis. Current evidence suggests that NRG1 fusion-positive NSCLC does not respond well to conventional treatments such as immunotherapy and chemotherapy. This review focuses on the biology and detection of NRG1 fusions and the evolving therapeutic landscape of NSCLC harboring NRG1 fusions. RECENT FINDINGS Zenocutuzumab, a bispecific antibody targeting HER2 and HER3, is the first FDA approved treatment for previously treated NRG1 fusion-positive NSCLC and pancreatic cancer. Additional NRG1 fusion directed strategies are in development. NRG1 fusions are rare molecular drivers of NSCLC that can be effectively treated with targeted therapies. Here, we summarize the biology and detection of NRG1 fusions, the currently approved bispecific antibody used to treat NRG1 fusion-positive NSCLC, and new agents under investigation.
Collapse
Affiliation(s)
- Brinda Gupta
- Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road NW, Washington, DC, 20007, USA
| | | | - Stephen V Liu
- Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road NW, Washington, DC, 20007, USA.
| |
Collapse
|
22
|
Wang L, You Y, He W, Hou Y, Li L, Wang L, Jiang C, Yi J, Xia Y, Xia L. Previous treatment decreases efficacy of pralsetinib in RET fusion-positive non-small-cell lung cancer. Front Med (Lausanne) 2025; 12:1467871. [PMID: 39926433 PMCID: PMC11804255 DOI: 10.3389/fmed.2025.1467871] [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: 07/21/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Background Pralsetinib is a selective RET inhibitor. The ARROW trial revealed that RET fusion-positive non-small-cell lung cancer (NSCLC) can benefit from pralsetinib with tolerable adverse events (AEs). However, the efficacy and safety of pralsetinib in real world has rarely been reported. Materials and methods This study reviewed the efficacy and safety of pralsetinib in RET fusion-positive NSCLC patients between March 2021 and December 2021. Progression-free survival (PFS) and overall survival (OS) were evaluated by a Kaplan-Meier analysis and log-rank test. A Cox regression model was performed to identify independent prognostic factors. Results A total of 28 patients were enrolled, and the median follow-up time was 18.1 months. The objective response rate and disease control rate of the whole cohort were 57.2% and 71.4%, respectively, and the median PFS and OS were 8.1 months [95% confidence interval (CI), 3.1-13.2] and 13.8 months (95% CI, 2.8-24.8), respectively. Baseline characteristics of the treatment naive group and pre-treated group were listed. The median PFS tended to be better in treatment naive group (18.3 vs. 8.0 months, P = 0.067), while the median OS were similar between the two groups (28.4 vs. 11.6 months, P = 0.308). Patients with Eastern Cooperative Oncology Group Performance Status (ECOG PS) score of 2 had worse median PFS comparing with those with ECOG PS score of 0-1 (3.8 vs. 12.6 months, P = 0.004). Besides, patients previously treated with platinum-based chemotherapy (PBC) also revealed worse median PFS comparing with those without previous PBC (8.0 vs. 18.6 months, P = 0.023). Furthermore, patients previously treated with anti-programmed death-1 (PD-1) antibody or multikinase inhibitors (MKIs) showed worse median OS compared with those without previous anti-PD-1 antibody (5.0 vs. 22.0 months, P = 0.002) or MKIs (6.2 vs. 28.4 months, P = 0.015). The most common AEs was increased aspartate aminotransferase (39.3%). Conclusion Pralsetinib was effective in RET fusion-positive NSCLC with tolerable AEs in real-world practice. Efficacy of pralsetinib was decreased in patients previously treated with PBC, immunotherapy, or MKIs.
Collapse
Affiliation(s)
- Lei Wang
- Department of Radiotherapy, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Pecking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Yafei You
- The Department of Clinical Oncology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Wenzhuo He
- Department of VIP Region, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Yu Hou
- Department of Radiotherapy, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Pecking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Lan Li
- Department of Radiotherapy, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Pecking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Li Wang
- Department of Radiotherapy, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Pecking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Chang Jiang
- Department of VIP Region, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Jiahong Yi
- Department of VIP Region, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Yaoxiong Xia
- Department of Radiotherapy, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Pecking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Liangping Xia
- Department of VIP Region, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
23
|
Mina SA, Shanshal M, Leventakos K, Parikh K. Emerging Targeted Therapies in Non-Small-Cell Lung Cancer (NSCLC). Cancers (Basel) 2025; 17:353. [PMID: 39941723 PMCID: PMC11816067 DOI: 10.3390/cancers17030353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/13/2025] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Targeted therapies have changed the treatment landscape of non-small-cell lung cancer and led to improved patient survival across all stages of lung cancer. Newer advances in common and novel oncogenic drivers continue to occur at vigorous speed, making it challenging to stay up to date with the rapidly evolving field. In this article, we review the emerging perspectives in the treatment of actionable targets in lung cancer. We focus on the development of newer KRAS-directed therapies, particularly on non-G12C mutations, pan-RAS inhibitors, and RAS-GTP inhibitors. We also describe the current standard of care for EGFR- and ALK-altered NSCLC and dive into the novel treatments expected to be in the clinic soon. A similar approach is taken toward MET, HER2, RET, ROS1, and FGFR alterations as emerging targets in non-small-cell lung cancer. Finally, we conclude this review with the current body of evidence for targeting TROP-2 as a novel target, potentially of importance in post-targeted therapy scenarios.
Collapse
Affiliation(s)
- Syeda A. Mina
- Division of Hematology and Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Kaushal Parikh
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
24
|
Pagliaro R, Medusa PM, Vitiello F, Aronne L, Campbell SFM, Perrotta F, Bianco A. Case report: Selpercatinib in the treatment of RET fusion-positive advanced lung adenocarcinoma: a challenging clinical case. Front Oncol 2025; 14:1500449. [PMID: 39882443 PMCID: PMC11774735 DOI: 10.3389/fonc.2024.1500449] [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: 09/23/2024] [Accepted: 12/12/2024] [Indexed: 01/31/2025] Open
Abstract
Background Rearranged during transfection (RET) fusions represent a distinct molecular subset of non-small cell lung cancer (NSCLC) with targeted therapeutic potential. Selpercatinib, a highly selective RET inhibitor, has demonstrated efficacy in various solid tumors harboring RET alterations. Here, we present a case highlighting the use and clinical outcomes of selpercatinib in a patient diagnosed with advanced lung adenocarcinoma harboring a RET fusion. Case presentation A 59-year-old woman with a history of stage IV lung adenocarcinoma harboring a KIF5B-RET fusion presented with disease progression following first-line chemo-immunotherapy. Selpercatinib was initiated as a targeted therapy, leading to a notable radiographic response and clinical improvement. The patient experienced a significant reduction in tumor burden and reported improved symptom control, with no significant adverse effects during the 21-month follow-up period. Conclusions This case highlights the efficacy and tolerability of selpercatinib in treating advanced lung adenocarcinoma with a RET fusion. The observed clinical response supports the early use of selpercatinib as a targeted therapy for RET fusion-positive NSCLC, including in patients with compromised general and respiratory conditions, especially in cases refractory to conventional treatments. Long-term follow-up studies are warranted to validate these findings and assess the durability of responses.
Collapse
Affiliation(s)
- Raffaella Pagliaro
- Department of Translational Medical Sciences University of Campania L. Vanvitelli, Naples, Italy
- Clinic of Respiratory Diseases “Vanvitelli”, A.O. dei Colli, Monaldi Hospital, Naples, Italy
| | - Paola Maria Medusa
- Department of Translational Medical Sciences University of Campania L. Vanvitelli, Naples, Italy
- Department of Pneumology and Oncology, Monaldi Hospital A.O. Dei Colli, Naples, Italy
| | - Fabiana Vitiello
- Department of Pneumology and Oncology, Monaldi Hospital A.O. Dei Colli, Naples, Italy
| | - Luigi Aronne
- Department of Translational Medical Sciences University of Campania L. Vanvitelli, Naples, Italy
- Clinic of Respiratory Diseases “Vanvitelli”, A.O. dei Colli, Monaldi Hospital, Naples, Italy
| | - Susan F. M. Campbell
- Department of Translational Medical Sciences University of Campania L. Vanvitelli, Naples, Italy
- Clinic of Respiratory Diseases “Vanvitelli”, A.O. dei Colli, Monaldi Hospital, Naples, Italy
| | - Fabio Perrotta
- Department of Translational Medical Sciences University of Campania L. Vanvitelli, Naples, Italy
- Clinic of Respiratory Diseases “Vanvitelli”, A.O. dei Colli, Monaldi Hospital, Naples, Italy
| | - Andrea Bianco
- Department of Translational Medical Sciences University of Campania L. Vanvitelli, Naples, Italy
- Clinic of Respiratory Diseases “Vanvitelli”, A.O. dei Colli, Monaldi Hospital, Naples, Italy
| |
Collapse
|
25
|
Foffano L, Bertoli E, Bortolot M, Torresan S, De Carlo E, Stanzione B, Del Conte A, Puglisi F, Spina M, Bearz A. Immunotherapy in Oncogene-Addicted NSCLC: Evidence and Therapeutic Approaches. Int J Mol Sci 2025; 26:583. [PMID: 39859299 PMCID: PMC11765476 DOI: 10.3390/ijms26020583] [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: 12/20/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. The discovery of specific driver mutations has revolutionized the treatment landscape of oncogene-addicted NSCLC through targeted therapies, significantly improving patient outcomes. However, immune checkpoint inhibitors (ICIs) have demonstrated limited effectiveness in this context. Emerging evidence, though, reveals significant heterogeneity among different driver mutation subgroups, suggesting that certain patient subsets may benefit from ICIs, particularly when combined with other therapeutic modalities. In this review, we comprehensively examine the current evidence on the efficacy of immunotherapy in oncogene-addicted NSCLC. By analyzing recent clinical trials and preclinical studies, along with an overview of mechanisms that may reduce immunotherapy efficacy, we explored potential strategies to address these challenges, to provide insights that could optimize immunotherapy approaches and integrate them effectively into the treatment algorithm for oncogene-addicted NSCLC.
Collapse
Affiliation(s)
- Lorenzo Foffano
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Elisa Bertoli
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Martina Bortolot
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Sara Torresan
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Elisa De Carlo
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Brigida Stanzione
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Alessandro Del Conte
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Fabio Puglisi
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Michele Spina
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Alessandra Bearz
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| |
Collapse
|
26
|
Wang Y, Hu X, Pandey S, Khatri U, Shen T, Subbiah V, Mooers BHM, Chao T, Wang S, Yu H, Sun X, Wu J, Cai J. Targeting Oncogenic RET Kinase by Simultaneously Inhibiting Kinase Activity and Degrading the Protein. J Med Chem 2025; 68:81-94. [PMID: 39723919 DOI: 10.1021/acs.jmedchem.4c01424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
The rearranged-during-transfection (RET) kinase is a validated target for the treatment of RET-altered cancers. Currently approved RET-selective kinase inhibitors, selpercatinib (LOXO-292) and pralsetinib (BLU-667), increase the oncogenic RET protein level upon treatment, which may affect their efficacy. We seek to reduce the oncogenic RET protein level and RET kinase activity simultaneously. Here, we report the development of proteolysis targeting chimera (PROTAC) degraders of oncogenic RET protein. Compound YW-N-7 exhibited dual action of selectively inhibiting and depleting RET protein both in vitro and in vivo. Proteomic analysis indicated that YW-N-7 is highly specific to RET. In cell cultures, reducing RET fusion protein potentiated the activity of LOXO-292. Furthermore, YW-N-7 showed significant activity in inhibiting KIF5B-RET-driven xenograft tumors in animals. This study exemplifies the feasibility of simultaneously inhibiting and degrading oncogenic RET kinase for cancer therapy.
Collapse
Affiliation(s)
- Yafeng Wang
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Xueqing Hu
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Shriya Pandey
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Ujjwol Khatri
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Tao Shen
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Vivek Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute, Nashville, Tennessee 37203, United States
| | - Blaine H M Mooers
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Ting Chao
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Shaohui Wang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Huaxuan Yu
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Xingmin Sun
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Jie Wu
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| |
Collapse
|
27
|
Melosky B, Juergens RA, Banerji S, Sacher A, Wheatley-Price P, Snow S, Tsao MS, Leighl NB, Martins I, Cheema P, Liu G, Chu QSC. The continually evolving landscape of novel therapies in oncogene-driven advanced non-small-cell lung cancer. Ther Adv Med Oncol 2025; 17:17588359241308784. [PMID: 39776537 PMCID: PMC11705342 DOI: 10.1177/17588359241308784] [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: 10/09/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a highly heterogeneous disease that is frequently associated with a host of known oncogenic alterations. Advances in molecular diagnostics and drug development have facilitated the targeting of novel alterations such that the majority of NSCLC patients have driver mutations that are now clinically actionable. The goal of this review is to gain insights into clinical research and development principles by summary, analysis, and discussion of data on agents targeting known alterations in oncogene-driven, advanced NSCLC beyond those in the epidermal growth factor receptor (EGFR) and the anaplastic lymphoma kinase (ALK). A search of published and presented literature was conducted to identify prospective trials and integrated analyses reporting outcomes for agents targeting driver gene alterations (except those in EGFR and ALK) in molecularly selected, advanced NSCLC. Clinical efficacy data were extracted from eligible reports and summarized in text and tables. Findings show that research into alteration-directed therapies in oncogene-driven, advanced NSCLC is an extremely active research field. Ongoing research focuses on the expansion of new agents targeting both previously identified targets (particularly hepatocyte growth factor receptor (MET), human epidermal growth factor receptor 2 (HER2), and Kirsten rat sarcoma viral oncogene homolog (KRAS)) as well as novel, potentially actionable targets (such as neuregulin-1 (NRG1) and phosphatidylinositol 3-kinase (PI3K)). The refinement of biomarker selection criteria and the development of more selective and potent agents are allowing for increasingly specific and effective therapies and the expansion of clinically actionable alterations. Clinical advances in this field have resulted in a large number of regulatory approvals over the last 3 years. Future developments should focus on the continued application of alteration therapy matching principles and the exploration of novel ways to target oncogene-driven NSCLC.
Collapse
Affiliation(s)
- Barbara Melosky
- Medical Oncology, BC Cancer Agency—Vancouver, University of British Columbia, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | | | - Shantanu Banerji
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Paul Wheatley-Price
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Stephanie Snow
- QEII Health Sciences Centre, Dalhousie University, Halifax, NS, Canada
| | - Ming-Sound Tsao
- University Health Network and Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Natasha B. Leighl
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | | | - Parneet Cheema
- William Osler Health System, University of Toronto, Brampton, ON, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Quincy S. C. Chu
- Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
28
|
Malhotra J, Mambetsariev I, Gilmore G, Fricke J, Nam A, Gallego N, Chen BT, Chen M, Amini A, Lukas RV, Salgia R. Targeting CNS Metastases in Non-Small Cell Lung Cancer With Evolving Approaches Using Molecular Markers: A Review. JAMA Oncol 2025; 11:60-69. [PMID: 39602134 DOI: 10.1001/jamaoncol.2024.5218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Importance Central nervous system (CNS) metastases presenting as either brain parenchymal metastases or leptomeningeal metastases are diagnosed in up to 50% of patients with advanced non-small cell lung cancer during their disease course. While historically associated with a poor prognosis due to limited treatment options, the availability of an increasing number of targeted therapies with good CNS penetration has significantly improved clinical outcomes for these patients. This has occurred in parallel with a more nuanced understanding of prognostic factors. Observations Multiple clinical trials have reported that disease control can be observed with targeted therapies with adequate CNS penetration, particularly for patients with molecular alterations in EGFR, ALK, ROS1, and RET. For these tumors, systemic targeted therapy may be used first for the management of CNS metastases, prior to considering radiation therapy (RT). At the time of isolated progression in the CNS, RT may be considered for the progressing lesions with continuation of the same systemic therapy. For other molecular alterations as well as for patients treated with checkpoint inhibitors, data are not yet clear if systemic therapy is sufficient for untreated CNS metastases, and early RT may need to be integrated into the treatment planning. An increasing number of studies investigate the role that emerging techniques, such as the sequencing of tumor DNA from resected brain metastases tissue or cerebrospinal fluid or radiomics-based analysis of CNS imaging, can play in guiding treatment approaches. Conclusions and Relevance With multiple generations of targeted therapies now available, the treatment for CNS metastases should be tailored to the patients with consideration given to molecular testing results, CNS penetrance of systemic therapy, patient characteristics, and multidisciplinary review. More research is needed in understanding the clonal evolution of CNS metastases, and the development of novel therapeutics with CNS efficacy.
Collapse
Affiliation(s)
- Jyoti Malhotra
- City of Hope Comprehensive Cancer Center, Duarte, California
| | | | - Gregory Gilmore
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Jeremy Fricke
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Arin Nam
- University of California, San Diego
| | | | - Bihong T Chen
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Mike Chen
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Arya Amini
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Rimas V Lukas
- Robert H. Lurie Comprehensive Cancer Center at Northwestern University, Chicago, Illinois
| | - Ravi Salgia
- City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
29
|
Meng Z, Pan T, Yu J, Shi C, Liu X, Xue D, Wang J, Ma B. Burden of thyroid cancer in China and worldwide from 1990 to 2021: observation, comparison, and forecast from the Global Burden of Disease Study 2021. Front Endocrinol (Lausanne) 2024; 15:1500926. [PMID: 39713055 PMCID: PMC11658972 DOI: 10.3389/fendo.2024.1500926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/19/2024] [Indexed: 12/24/2024] Open
Abstract
Background Thyroid cancer (TC) is a prevalent malignant tumor of the endocrine system in China. Current research primarily focuses on clinical diagnosis and treatment as well as underlying mechanisms, lacking epidemiological studies on the burden of the disease in China and worldwide. Methods The Global Burden of Disease Study 2021 was utilized to assess the incidence, prevalence, death, and disability-adjusted life years of TC in China and worldwide from 1990 to 2021 using the Joinpoint and R software. Results From 1990 to 2021, the incidence and prevalence rates of TC in China have been consistently rising, and their growth rates are far higher than the global average. In China, TC usually occurs in patients aged 50-59, and the crude death rate generally increases with age. The burden of death among females has gradually declined, while that among males has continued to increase and surpassed females at the beginning of the 21st century. The burden of TC is heavy among middle-aged and elderly populations and the younger populations is also rapidly rising. The increased number of TC is mainly attributed to epidemiological changes, while the increase of deaths in China is primarily due to aging and population. Additionally, we predict that the age-standardized incidence rate of TC in China will continue to grow slowly over the next decade, while the age-standardized death rate will gradually decline among females and stabilize among males. Conclusion It is imperative to avoid over-screening and over-treatments for TC. Meanwhile, we should also avoid missing aggressive types of TC that may have an impact on overall survival. Additionally, understanding the mechanisms of metastasis and improving clinical treatments should be prioritized for further investigation. TC remains a significant public health challenge in China, necessitating a careful balance of the cost-benefit ratio.
Collapse
Affiliation(s)
- Ziang Meng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ti Pan
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingjing Yu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chao Shi
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuxu Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Wang
- NHC Key Laboratory of Etiology and Epidemiology (National Health Commission of the People’s Republic of China), Harbin Medical University, Harbin, China
| | - Biao Ma
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
30
|
Podder V, Ranjan T, Gowda M, Camacho AM, Ahluwalia MS. Emerging Therapies for Brain Metastases in NSCLC, Breast Cancer, and Melanoma: A Critical Review. Curr Neurol Neurosci Rep 2024; 25:6. [PMID: 39625633 DOI: 10.1007/s11910-024-01388-1] [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] [Accepted: 10/19/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE OF REVIEW Advancements in precision medicine have shifted the treatment paradigm of brain metastases (BM) from non-small cell lung cancer (NSCLC), breast cancer, and melanoma, especially through targeted therapies focused on specific molecular drivers. These novel agents have improved outcomes by overcoming challenges posed by the blood-brain barrier (BBB) and resistance mechanisms, enabling more effective treatment of BM. RECENT FINDINGS In NSCLC, therapies such as osimertinib have improved efficacy in treating EGFR-mutant BM, with emerging combinations such as amivantamab and lazertinib offering promising alternatives for patients resistant to frontline therapies. In HER2-positive breast cancer, significant advancements with tucatinib and trastuzumab deruxtecan (T-DXd) have transformed the treatment landscape, achieving improved survival and intracranial control in patients with BM. Similarly, in triple-negative breast cancer (TNBC), novel therapies such as sacituzumab govitecan (SG) and datopotamab deruxtecan (Dato-DXd) offer new hope for managing BM. For melanoma, the combination of immune checkpoint inhibitors such as nivolumab and ipilimumab has proven effective in enhancing survival for patients with BM, both in BRAF-mutant and wild-type cases. Developing targeted therapies penetrating the BBB has revolutionized BM treatment by targeting key drivers like EGFR, ALK, HER2, and BRAF. Despite improved survival, challenges persist, particularly for patients with resistant genetic alterations. Future research should optimise combination therapies, overcome resistance, and refine treatment sequencing. Continued emphasis on personalized, biomarker-driven approaches offers the potential to further improve outcomes, even for complex cases.
Collapse
Affiliation(s)
- Vivek Podder
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Tulika Ranjan
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Maya Gowda
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | | | | |
Collapse
|
31
|
Yi H, Cao Y, Shi F, Wei X, Han S. Cost-effectiveness analysis of selpercatinib versus chemotherapy and pembrolizumab in the first-line treatment of rearranged during transfection fusion-positive non-small cell lung cancer in the United States. Int J Clin Pharm 2024; 46:1427-1435. [PMID: 39352419 DOI: 10.1007/s11096-024-01800-3] [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: 04/29/2024] [Accepted: 08/26/2024] [Indexed: 10/03/2024]
Abstract
BACKGROUND Although selpercatinib has shown clinical benefits for rearranged during transfection (RET) fusion-positive non-small cell lung cancer (NSCLC), its cost-effectiveness requires further evaluation. AIM This study aimed to evaluate the cost-effectiveness of selpercatinib versus chemotherapy and pembrolizumab in the first-line treatment of RET fusion-positive NSCLC from the perspective of the United States (US) payer. METHOD A partitioned survival model was developed based on data from the LIBRETTO-431 trial. Cost and utility values for the health state were obtained from database data or published literature. The measured outcomes included quality-adjusted life-years (QALYs) and the incremental cost-effectiveness ratio (ICER). One-way sensitivity analysis and probabilistic sensitivity analyses (PSA) were conducted to assess the uncertainty of the model. RESULTS Selpercatinib increased QALYs in patients with RET fusion-positive NSCLC by 0.90 compared to chemotherapy plus pembrolizumab, with an additional cost of $542,517.45, resulting in an ICER of $603,286.49/QALY, which exceeded the willingness-to-pay (WTP) threshold ($150,000) in the US. One-way sensitivity analysis suggested that the utility of progressed disease, the utility of progression-free survival, the price of selpercatinib, the discount, the price of pemetrexed, and the price of pembrolizumab had the greatest influence on the cost- effectiveness analysis process. In the PSA, 99.9% of the scatter points were distributed above the US WTP threshold of $150,000. CONCLUSION From the perspective of the US payer, selpercatinib is not cost-effective compared to chemotherapy and pembrolizumab for first-line treatment in patients with RET fusion-positive NSCLC.
Collapse
Affiliation(s)
- Hongbin Yi
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- International Research Center for Medicinal Administration, Peking University, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Yingdan Cao
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- International Research Center for Medicinal Administration, Peking University, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Fenghao Shi
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- International Research Center for Medicinal Administration, Peking University, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Xiaoxia Wei
- Department of Pharmacy, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, 350001, China.
| | - Sheng Han
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- International Research Center for Medicinal Administration, Peking University, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| |
Collapse
|
32
|
Podder V, Bellur S, Margolin K, Advani P, Mahtani RL, Subbiah V, Novo GB, Ranjan T, Ahluwalia MS. Immunotherapeutic and Targeted Strategies for Managing Brain Metastases from Common Cancer Origins: A State-of-the-Art Review. Curr Oncol Rep 2024; 26:1612-1638. [PMID: 39514054 DOI: 10.1007/s11912-024-01593-8] [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] [Accepted: 08/01/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW This review examines contemporary strategies for managing brain metastases (BM) from common cancers such as lung, breast, and melanoma. We evaluate the efficacy and applicability of targeted therapies and immunotherapies, exploring their potential to cross the blood-brain barrier and improve patient outcomes. RECENT FINDINGS Recent studies have shown that tyrosine kinase inhibitors, immune checkpoint inhibitors, and ADCs effectively treat BM. These treatments can overcome the challenges posed by the blood-brain barrier and improve therapeutic outcomes. ADCs are promising because they can deliver cytotoxic agents directly to tumor cells, which reduces systemic toxicity and increases drug delivery efficiency to the brain. Personalized medicine is becoming increasingly significant in treatment decisions, with biomarkers playing an essential role. Advances in molecular genetics and drug development have led to more refined treatments, emphasizing the precision medicine framework. The management of BM is evolving, driven by drug efficacy, resistance mechanisms, and the need for personalized medicine. Integrating ADCs into treatment regimens represents a significant advancement in targeting metastatic brain tumors. Despite these advances, BM management still presents considerable challenges, requiring ongoing research and multi-institutional trials to optimize therapeutic strategies. This review outlines the current state and future directions in treating BM, highlighting the critical need for continued innovation and comprehensive clinical evaluations to improve survival rates and quality of life for affected patients.
Collapse
Affiliation(s)
- Vivek Podder
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Shreyas Bellur
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Kim Margolin
- Saint John's Cancer Institute, Santa Monica, CA, USA
| | | | - Reshma L Mahtani
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Vivek Subbiah
- Sarah Cannon Research Institute (SCRI), Nashville, TN, USA
| | - Gabriella B Novo
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Tulika Ranjan
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | | |
Collapse
|
33
|
Jo Y, Chipman JJ, Haaland B, Greene T, Kohli M. Multigene Copy Number Alteration Risk Score Biomarker-Based Enrichment Study Designs in Metastatic Castrate-Resistant Prostate Cancer. JCO Precis Oncol 2024; 8:e2400399. [PMID: 39626157 PMCID: PMC11627311 DOI: 10.1200/po-24-00399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/05/2024] [Accepted: 10/17/2024] [Indexed: 12/11/2024] Open
Abstract
PURPOSE A composite multigene risk score derived from tumor-biology alterations specific to metastatic castrate-resistant prostate cancer (mCRPC) state was evaluated as a classifier to design biomarker-based enrichment clinical trials. METHODS A plasma cell-free DNA copy number alteration risk score based on alterations in 24 genes was simulated to develop a biomarker classifier-based clinical trial design enriched for high-risk patients to detect a survival advantage of a novel treatment (hazard ratio of 0.70 with 80% power). We determined the design trade-offs between the number of patients screened and enrolled when varying the type of patients to enrich and the extent of enrichment needed. RESULTS For a 2-year overall survival end point in mCRPC state, fully enriching patients with mCRPC having a high-risk score of 3 or more (the 95th percentile of a range of risk scores in patients with mCRPC) was determined to require screening to a maximum of 4,149 patients to enroll 259 patients for the targeted effect size. A nonenriched trial was determined to require enrolling 689 patients to be equivalently powered. We identified a pragmatic alternative, which is to enrich patients with mCRPC with a risk score of 1 or more (the 67th percentile) and an enrichment fraction of 0.25. This would require screening 658 patients to enroll 584 patients, and it maximizes the ability to detect a difference in treatment effect by risk score. CONCLUSION A plasma multi-CNA risk score classifier can feasibly be leveraged to design an enrichment trial in mCRPC. Enriching 25% of patients screened with a risk score >1 was observed to be optimal for obtaining an adequately powered, biomarker-based mCRPC-enriched clinical trial.
Collapse
Affiliation(s)
- Yeonjung Jo
- Division of Biostatistics, Department of Population Health Sciences, School of Medicine, University of Utah, Salt Lake City, UT
- Cancer Biostatistics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Jonathan J. Chipman
- Division of Biostatistics, Department of Population Health Sciences, School of Medicine, University of Utah, Salt Lake City, UT
- Cancer Biostatistics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | | | - Tom Greene
- Division of Biostatistics, Department of Population Health Sciences, School of Medicine, University of Utah, Salt Lake City, UT
- Cancer Biostatistics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Manish Kohli
- Division of Oncology, Department of Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
34
|
Sun Y, Ma L, Zhang X, Wang Z. Advances in the Treatment of Rare Mutations in Non-Small Cell Lung Cancer. Onco Targets Ther 2024; 17:1095-1115. [PMID: 39583247 PMCID: PMC11585992 DOI: 10.2147/ott.s487870] [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: 07/20/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024] Open
Abstract
Lung cancer is a malignant tumor with the highest morbidity and mortality rate worldwide, with nearly 2.5 million new cases and more than 1.8 million deaths reported globally in 2022. Lung cancer is broadly categorized into two main types: non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC), with NSCLC accounting for about 85% of all cases. Early-stage lung cancers often present without obvious symptoms, resulting in most patients being diagnosed at an advanced stage where traditional chemotherapy has limited efficacy. Recent advances in molecular biology have elucidated the pivotal role of gene mutations in tumor development, paving the way for targeted therapies that have markedly benefited patients. Beyond the well-known epidermal growth factor receptor (EGFR) mutation, an increasing number of new molecular targets have been identified, including ROS1 rearrangement, BRAF mutation, NTRK fusion, RET fusion, MET mutation, KRAS G12C mutation, HER2 mutation, ALK rearrangement, and NRG1 fusion. Some of these targeted therapies have already been approved by the Food and Drug Administration (FDA), and many others are currently undergoing clinical trials. This review summarizes recent advances in NSCLC treatment with molecular targets, highlighting progress, challenges, and their impact on patient prognosis.
Collapse
Affiliation(s)
- Yanning Sun
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Li Ma
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Xiaofei Zhang
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Zhaoxia Wang
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
35
|
Yin Y, Sun F, Jin Y. A real-world pharmacovigilance study of FDA Adverse Event Reporting System events for pralsetinib. Front Oncol 2024; 14:1491167. [PMID: 39600647 PMCID: PMC11588741 DOI: 10.3389/fonc.2024.1491167] [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: 09/04/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Background Pralsetinib, a selective oral inhibitor of rearranged during transfection (RET) fusion proteins and oncogenic RET mutants, has shown significant efficacy in treating RET fusion-positive non-small cell lung cancer and thyroid cancer. However, since pralsetinib was approved in the United States in September 2020, there have been limited reports of post-marketing adverse events (AEs). In this study, we aimed to analyze the AE signals with pralsetinib on the basis of the United States Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) to provide instructions in clinical practice. Methods All AE reports were obtained from the FAERS database from the first quarter (Q3) of 2020 to the second quarter (Q2) of 2024. Various signal quantification techniques were used for analysis, including reporting odds ratios, proportional reporting ratios, Bayesian confidence propagation neural network, and multi-item gamma Poisson shrinker (MGPS)-based empirical Bayesian geometric mean. Results Out of 8,341,673 case reports in the FAERS database, 1,064 reports of pralsetinib as the "primary suspected (PS)" AEs were recorded, covering 26 system organ classes and 256 preferred terms. Of the reports, 62.5% were from consumers rather than healthcare professionals. The most common systems were general disorders and administration site conditions (n = 704), investigations (n = 516), and gastrointestinal disorders (n = 405). A total of 95 significant disproportionality preferred terms (PTs) conformed to the four algorithms simultaneously. AEs that ranked the top three at the PT level were hypertension (n = 80), asthenia (n = 79), and anemia (n = 65). Of the 95 PTs with significant disproportionation, unexpected significant AEs such as increased blood calcitonin, increased myocardial necrosis marker, and bacterial cystitis were observed, which were not mentioned in the drug's instructions. The median onset time of pralsetinib-associated AEs was 41 days [interquartile range (IQR) 14-86 days]. The majority of the AEs occurred in 30 days (42.86%). Conclusion Our pharmacovigilance analysis of real-world data from the FEARS database revealed the safety signals and potential risks of pralsetinib usage. These results can provide valuable evidence for further clinical application of pralsetinib and are important in enhancing clinical medication safety.
Collapse
Affiliation(s)
- Yi Yin
- Department of Pediatric Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Fengli Sun
- Department of Pediatric Intensive Care Unit, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Youpeng Jin
- Department of Pediatric Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
36
|
Hockemeyer KG, Rusthoven CG, Pike LRG. Advances in the Management of Lung Cancer Brain Metastases. Cancers (Basel) 2024; 16:3780. [PMID: 39594735 PMCID: PMC11593022 DOI: 10.3390/cancers16223780] [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: 10/03/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Lung cancer, both non-small cell and small cell, harbors a high propensity for spreading to the central nervous system. Radiation therapy remains the backbone of the management of brain metastases. Recent advances in stereotactic radiosurgery have expanded its indications and ongoing studies seek to elucidate optimal fractionation and coordination with systemic therapies, especially targeted inhibitors with intracranial efficacy. Efforts in whole-brain radiotherapy aim to preserve neurocognition and to investigate the need for prophylactic cranial irradiation. As novel combinatorial strategies are tested and prognostic/predictive biomarkers are identified and tested, the management of brain metastases in lung cancer will become increasingly personalized to optimally balance intracranial efficacy with preserving neurocognitive function and patient values.
Collapse
Affiliation(s)
- Kathryn G. Hockemeyer
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chad G. Rusthoven
- Department of Radiation Oncology, University of Colorado, Aurora, CO 80045, USA
| | - Luke R. G. Pike
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
37
|
Owen D, Ben-Shachar R, Feliciano J, Gai L, Beauchamp KA, Rivers Z, Hockenberry AJ, Harrison G, Guittar J, Catela C, Parsons J, Cohen E, Sasser K, Nimeiri H, Guinney J, Patel J, Morgensztern D. Actionable Structural Variant Detection via RNA-NGS and DNA-NGS in Patients With Advanced Non-Small Cell Lung Cancer. JAMA Netw Open 2024; 7:e2442970. [PMID: 39495511 PMCID: PMC11536281 DOI: 10.1001/jamanetworkopen.2024.42970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 09/12/2024] [Indexed: 11/05/2024] Open
Abstract
Importance The National Comprehensive Cancer Network (NCCN) guidelines for non-small cell lung cancer suggest that RNA next-generation sequencing (NGS) may improve the detection of fusions and splicing variants compared with DNA-NGS alone. However, there is limited adoption of RNA-NGS in routine oncology clinical care today. Objective To analyze clinical evidence from a diverse cohort of patients with advanced lung adenocarcinoma and compare the detection of NCCN-recommended actionable structural variants (aSVs; fusions and splicing variants) via concurrent DNA and RNA-NGS vs DNA-NGS alone. Design, Setting, and Participants This multisite, retrospective cohort study examined patients sequenced between February 2021 and October 2023 within the deidentified, Tempus multimodal database, consisting of linked molecular and clinical data. Participants included patients with advanced lung adenocarcinoma and sufficient tissue sample quantities for both RNA-NGS and DNA-NGS testing. Exposures Received results from RNA-NGS and DNA-NGS solid-tissue profiling assays. Main Outcomes and Measures Detection rates of NCCN guideline-based structural variants (ALK, ROS1, RET and NTRK1/2/3 fusions, as well as MET exon 14 skipping splicing alterations) found uniquely by RNA-NGS. Results In the evaluable cohort of 5570 patients, median (IQR) age was 67.8 (61.3-75.4) years, and 2989 patients (53.7%) were female. The prevalence of actionable structural variants detected by either RNA-NGS or DNA-NGS was 8.8% (n = 491), with 86.7% (n = 426) of these detected by DNA-NGS. Concurrent RNA-NGS and DNA-NGS identified 15.3% more patients harboring aSVs compared with DNA-NGS alone (491 vs 426 patients, respectively), including 14.3% more patients harboring actionable fusions (376 vs 329 patients) and 18.6% more patients harboring MET exon 14 skipping alterations (115 vs 97 patients). There was no significant association between the assay used for aSV detection and aSV-targeted therapeutic adoption or clinical outcome. Emerging structural variants (eSVs) were found to have a combined prevalence to be 0.7%, with only 47.5% of eSVs detected by DNA-NGS. Conclusions and Relevance In this cohort study, the detection of structural variants via concurrent RNA-NGS and DNA-NGS was higher across multiple NCCN-guideline recommended biomarkers compared with DNA-NGS alone, suggesting that RNA-NGS should be routinely implemented in the care of patients with advanced NSCLC.
Collapse
Affiliation(s)
- Dwight Owen
- Ohio State University School of Medicine, Columbus
| | | | | | - Lisa Gai
- Tempus AI Inc, Chicago, Illinois
| | | | | | | | | | | | | | | | | | | | | | | | - Jyoti Patel
- Northwestern University School of Medicine, Chicago, Illinois
| | | |
Collapse
|
38
|
Wang A, Li T, Mao YY, Gao M, Shu S, Xia CH, Dong Y, Liu M, Wang JL, Ma JX, Hu Y. Evolution of treatment strategies for solid tumors with RET rearrangement in China and real-world treatment status of Non-small Cell Lung Cancer (NSCLC). BMC Pulm Med 2024; 24:552. [PMID: 39497173 PMCID: PMC11533424 DOI: 10.1186/s12890-024-03371-5] [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: 09/10/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024] Open
Abstract
OBJECTIVE The present study endeavors to furnish an exhaustive review of the research advancements on solid tumors harboring RET rearrangement within the Chinese context, particularly emphasizing the examination of real-world therapeutic strategies and clinical outcomes observed in individuals diagnosed with advanced non-small cell lung cancer (NSCLC). The review delves into a critical assessment of the therapeutic efficacy of targeted RET inhibitors, while also scrutinizing the diverse array of treatment modalities employed in the Chinese patient population. METHODS The study conducted a comprehensive review of the advancements made by Chinese scholars in the realm of RET driver genes. It delved into the analysis of the incidence of RET rearrangements in solid tumors, alongside an examination of the varied treatment paradigms and their current status within China. Utilizing the RECIST 1.1 criteria, the study evaluated the therapeutic efficacy achieved in RET-positive NSCLC patients undergoing diverse treatment modalities. Furthermore, treatment-related adverse events (TRAEs) were meticulously graded following the Common Terminology Criteria for Adverse Events (CTCAE). RESULTS A retrospective, multi-center, real-world analysis was conducted, encompassing 64 patients diagnosed with pathologically confirmed RET rearrangement advanced non-small cell lung cancer (NSCLC) between December 2015 and November 2023. Notably, KIF5B-RET emerged as the most prevalent RET fusion partner, accounting for 59.4% of cases. Therapeutic interventions among these patients included specific targeted inhibitors such as Pralsetinib (48.4%), chemotherapy (34.3%), multi-target inhibitors (15.6%), and one case (1.6%) involving immunotherapy combined with anti-angiogenic therapy. In terms of progression-free survival (PFS), Pralsetinib monotherapy demonstrated a median PFS of 16.03 months, outperforming chemotherapy (2.87 months; p < 0.0001), chemotherapy combined with anti-angiogenic therapy (6.90 months; p = 0.048), and multi-target inhibitors (2.50 months; p < 0.0001). Furthermore, the one-year and two-year overall survival (OS) rates for Pralsetinib monotherapy were 64.3% and 46.4%, respectively. Regarding safety, 71.0% of patients receiving Pralsetinib experienced at least one adverse event, with 45.2% classified as grade 3-4 in severity. Notably, no fatalities were attributed to adverse events. Common adverse events included hemoglobin reduction (35.5%) and neutropenia (32.3%), indicative of an overall favorable safety profile for Pralsetinib in this patient population. CONCLUSION This study encapsulates the research endeavors and treatment advancements of RET rearrangement solid tumors within the Chinese healthcare landscape, specifically highlighting the diverse real-world therapeutic approaches and their effectiveness in managing advanced RET rearrangement NSCLC among Chinese patients. Notably, targeted RET inhibitors like Pralsetinib have emerged as potent therapeutic agents, exhibiting remarkable efficacy and a manageable safety profile in this patient cohort. These findings underscore the potential of Pralsetinib and similar targeted therapies as novel treatment options for individuals with RET fusion-positive NSCLC.
Collapse
Affiliation(s)
- An Wang
- Department of Oncology, the First Medical Center of PLA General Hospital, (Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education, China)), Beijing, 100853, China
- Graduate School, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, 100853, China
| | - Tao Li
- Department of Oncology, the First Medical Center of PLA General Hospital, (Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education, China)), Beijing, 100853, China
- Graduate School, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, 100853, China
- Department of Stomatology, The First Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Yun-Ye Mao
- Department of Oncology, the First Medical Center of PLA General Hospital, (Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education, China)), Beijing, 100853, China
- Graduate School, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, 100853, China
| | - Ming Gao
- Department of Oncology, the First Medical Center of PLA General Hospital, (Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education, China)), Beijing, 100853, China
- Graduate School, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, 100853, China
| | - Sheng Shu
- Department of Oncology, the First Medical Center of PLA General Hospital, (Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education, China)), Beijing, 100853, China
- Graduate School, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, 100853, China
| | - Chang-Hong Xia
- Graduate School, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, 100853, China
- Department of Medical Ultrasonics, The Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Yi Dong
- Department of Oncology, the First Medical Center of PLA General Hospital, (Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education, China)), Beijing, 100853, China
- Graduate School, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, 100853, China
| | - Min Liu
- Graduate School, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, 100853, China
- Department of Pharmacy, Medical Supplies Center, the First Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Jin-Liang Wang
- Graduate School, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, 100853, China.
- Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100853, China.
| | - Jun-Xun Ma
- Graduate School, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, 100853, China.
- Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100853, China.
| | - Yi Hu
- Department of Oncology, the First Medical Center of PLA General Hospital, (Chinese PLA Key Laboratory of Oncology, Key Laboratory for Tumor Targeting Therapy and Antibody Drugs (Ministry of Education, China)), Beijing, 100853, China.
- Graduate School, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, 100853, China.
| |
Collapse
|
39
|
Li W, Wang Y, Xiong A, Gao G, Song Z, Zhang Y, Huang D, Ye F, Wang Q, Li Z, Liu J, Xu C, Sun Y, Liu X, Zhou F, Zhou C. First-in-human, phase 1 dose-escalation and dose-expansion study of a RET inhibitor SY-5007 in patients with advanced RET-altered solid tumors. Signal Transduct Target Ther 2024; 9:300. [PMID: 39489747 PMCID: PMC11532403 DOI: 10.1038/s41392-024-02006-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/12/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Oncogenic RET alteration is an important, tissue-agnostic therapeutic target across diverse cancers. We conducted a first-in-human phase 1 study on SY-5007, a potent and selective RET inhibitor, in patients with RET-altered solid tumors. Primary endpoints were safety, maximum tolerated dose (MTD), and recommended phase 2 dose (RP2D). Secondary endpoints included pharmacokinetics and preliminary anti-tumor activity. A total of 122 patients were enrolled (17 in dose-escalation phase and 105 in dose-expansion phase), including 91 with non-small cell lung cancer, 23 with medullary thyroid cancer, 7 with papillary thyroid cancer and 1 with gastric cancer. Treatment-related adverse events (TRAEs) were reported in 96.7% of patients, with the most common grade ≥ 3 TRAEs being hypertension (22.1%), diarrhea (16.4%), hypertriglyceridemia (6.6%), and neutropenia (6.6%). The exposure to SY-5007 was dose proportional. Among the 116 efficacy-evaluable patients, the overall objective response rate (ORR) was 57.8%, with 70.0% in treatment-naïve patients and 51.3% in previously treated patients. The median progression-free survival (PFS) was 21.1 months. Efficacy was observed regardless of tumor types and previous therapies. Biomarker analysis of 61 patients with circulating tumor DNA (ctDNA)-detectable RET alterations showed an ORR of 57.4% and median PFS of 13.8 months. Rapid ctDNA clearance of RET alteration correlated with faster responses and improved outcomes. In relapsed patients, off-target induced resistance was observed in 57.1% (12/21), with no on-target RET alterations identified. In conclusion, SY-5007 was well-tolerated and showed promising efficacy in patients with RET-altered solid tumors. Serial ctDNA monitoring may unveil treatment response and potential resistance mechanisms (NCT05278364).
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins c-ret/genetics
- Proto-Oncogene Proteins c-ret/antagonists & inhibitors
- Male
- Female
- Middle Aged
- Aged
- Adult
- Thyroid Neoplasms/drug therapy
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/pathology
- Carcinoma, Neuroendocrine/drug therapy
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Thyroid Cancer, Papillary/drug therapy
- Thyroid Cancer, Papillary/genetics
- Thyroid Cancer, Papillary/pathology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Neoplasms/drug therapy
- Neoplasms/genetics
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/genetics
- Stomach Neoplasms/pathology
- Maximum Tolerated Dose
- Protein Kinase Inhibitors/pharmacokinetics
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/pharmacology
- Aged, 80 and over
Collapse
Affiliation(s)
- Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yongsheng Wang
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Anwen Xiong
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Ge Gao
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Zhengbo Song
- Department of Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yiping Zhang
- Department of Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Dingzhi Huang
- Lung Cancer Department, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Feng Ye
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, Fujian, China
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Zhihui Li
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Jiaye Liu
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Chunwei Xu
- Department of Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yinghui Sun
- Shouyao Holdings (Beijing) Co., Ltd, Beijing, China
| | - Xijie Liu
- Shouyao Holdings (Beijing) Co., Ltd, Beijing, China
| | - Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
40
|
Chan KL, Faiz SA, Altan M, Sheshadri A. Updates in Drug-Related Pneumonitis Due to Targeted Oncologic Therapies. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2024; 7:272-282. [PMID: 39524467 PMCID: PMC11541925 DOI: 10.36401/jipo-24-12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/11/2024] [Accepted: 07/07/2024] [Indexed: 11/16/2024]
Abstract
An increasing number of newer targeted oncologic therapies approved for clinical use can cause drug-related pneumonitis. Drug-related pneumonitis can be difficult to diagnose and requires a high index of suspicion. This review serves as an update to a prior review in this journal about pneumonitis with precision oncology therapies. In this review, we focus on the incidence, timing of onset, and imaging patterns of pneumonitis associated with a number of newly approved precision oncologic agents, with a particular focus on new antibody-drug conjugate therapies.
Collapse
Affiliation(s)
- Kathy L. Chan
- Divisions of Critical Care, Pulmonary and Sleep Medicine, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Saadia A. Faiz
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mehmet Altan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
41
|
Wei X, Uchibori K, Kondo N, Utsumi T, Takemoto A, Koike S, Takagi S, Yanagitani N, Nishio M, Katayama R. MIG6 loss increased RET inhibitor tolerant persister cells in RET-rearranged non-small cell lung cancer. Cancer Lett 2024; 604:217220. [PMID: 39244004 DOI: 10.1016/j.canlet.2024.217220] [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: 04/25/2024] [Revised: 08/20/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Recently approved RET tyrosine kinase inhibitors (TKIs) have shown promising therapeutic effects against RET-rearranged non-small cell lung cancer (NSCLC) or RET-mutated thyroid cancer. However, resistance develops, limiting long-term efficacy. Although many RET-TKI resistance mechanisms, such as secondary mutations in RET or activation of bypass pathways, are known, some primary or acquired resistance mechanisms are unclear. Here, human genome-wide CRISPR/Cas9 screening was performed to identify genes related to drug-tolerant persister cells. Patient-derived cells with RET-fusion were introduced genome-wide sgRNA library and treated with RET-TKI for 9 days, resulting in the discovery of several candidate genes. Knockout of MED12 or MIG6 significantly increased residual drug-tolerant persister cells under RET-TKI treatment. MIG6 loss induced significant EGFR activation even with low concentrations of EGFR ligands and led to resistance to RET-TKIs. EGFR inhibition with afatinib or cetuximab in combination with RET TKIs was effective in addressing drug persistence. By contrast, a KIF5B-RET positive cells established from a RET-rearranged NSCLC patient, showed significant resistance to RET-TKIs and high dependence on EGFR bypass signaling. Consistently, knocking out EGFR or RET led to high sensitivity to RET or EGFR inhibitor respectively. Here, we have provided a comprehensive analysis of adaptive and acquired resistance against RET-rearranged NSCLC.
Collapse
Affiliation(s)
- Xinzhao Wei
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Ken Uchibori
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan; Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Nobuyuki Kondo
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan; Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takahiro Utsumi
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan; Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ai Takemoto
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Sumie Koike
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoshi Takagi
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Noriko Yanagitani
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makoto Nishio
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
42
|
Kojima M, Wende W, Zhao H. Tips for Accelerating BOIN Design. Ther Innov Regul Sci 2024; 58:1129-1137. [PMID: 39222294 DOI: 10.1007/s43441-024-00692-9] [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: 03/04/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
During discussions at the Data Science Roundtable meeting in Japan, there were instances where the adoption of the BOIN design was declined, attributed to the extension of study duration and increased sample size in comparison to the 3 + 3 design. We introduce an accelerated BOIN design aimed at completing a clinical phase I trial at a pace comparable to the 3 + 3 design. Additionally, we introduce how we could have applied the BOIN design within our company, which predominantly utilized the 3 + 3 design for most of its clinical oncology dose escalation trials. The accelerated BOIN design is adaptable by using efficiently designated stopping criterion for the existing BOIN framework. Our approach is to terminate the dose escalation study if the number of evaluable patients treated at the current dose reaches 6 and the decision is to stay at the current dose for the next cohort of patients. In addition, for lower dosage levels, considering a cohort size smaller than 3 may be feasible when there are no safety concerns from non-clinical studies. We demonstrate the accelerated BOIN design using a case study and subsequently evaluate the performance of our proposed design through a simulation study. In the simulation study, the average difference in the percentage of correct MTD selection between the accelerated BOIN design and the standard BOIN design was - 2.43%, the average study duration and the average sample size of the accelerated BOIN design was reduced by 14.8 months and 9.22, respectively, compared with the standard BOIN design.
Collapse
Affiliation(s)
- Masahiro Kojima
- Biometrics Department, Research Division, Kyowa Kirin Co., Ltd. Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo, 100-004, Japan.
| | - Wu Wende
- Biometrics Department, Development Division, Kyowa Kirin, Inc, Princeton, NJ, USA
| | - Henry Zhao
- Biometrics Department, Development Division, Kyowa Kirin, Inc, Princeton, NJ, USA
| |
Collapse
|
43
|
Koban MU, Hartmann M, Amexis G, Franco P, Huggins L, Shah I, Karachaliou N. Targeted Therapies, Novel Antibodies, and Immunotherapies in Advanced Non-Small Cell Lung Cancer: Clinical Evidence and Drug Approval Patterns. Clin Cancer Res 2024; 30:4822-4833. [PMID: 39177967 PMCID: PMC11528205 DOI: 10.1158/1078-0432.ccr-24-0741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/23/2024] [Accepted: 07/11/2024] [Indexed: 08/24/2024]
Abstract
Since 2011, the US FDA has approved 30 new drugs for use in advanced non-small cell lung cancer (NSCLC), mainly comprising tyrosine kinase inhibitors and immune checkpoint inhibitors. NSCLC with oncogene driver alterations is amenable to treatment with targeted drugs, usually small-molecule inhibitors. In these cases, the demonstration of high overall response rates, coupled with a lasting duration of response, has allowed for accelerated approval in the United States, based on single-cohort or multicohort trials. Confirmatory clinical evidence was subsequently provided through postmarketing trials. In NSCLC without such driver alterations, regulatory agencies in both the United States and the European Union set clinical evidence expectations that foster the conduct of studies primarily focused on determining survival or event-free survival, based on randomized controlled trial designs. This review analyzes the approval patterns of novel therapeutics for NSCLC with a focus on small-molecule inhibitors that target driver alterations, as well as biologics. The latter include mAbs inhibiting immune checkpoints like PD-(L)1 or cell surface receptors and antibody-drug conjugates, highly potent biologics linked to a cytotoxic compound. The differentiation of NSCLC into oncogene- and non-oncogene-addicted subtypes determines drug development strategies, the extent of the clinical development program, access to orphan drug development incentives, and regulatory approval strategies.
Collapse
Affiliation(s)
- Marén U. Koban
- Global Regulatory and Scientific Policy, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | - Georgios Amexis
- Global Regulatory Affairs Oncology, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Pedro Franco
- Merck Serono Limited UK, an Affiliate of Merck KGaA, Feltham, United Kingdom
| | - Laura Huggins
- Global Regulatory Affairs Oncology, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | - Niki Karachaliou
- Global Clinical Development, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| |
Collapse
|
44
|
Sampat PJ, Cortese A, Goodman A, Ghelani GH, Mix MD, Graziano S, Basnet A. Treatment of brain metastases from non-small cell lung cancer: preclinical, clinical, and translational research. Front Oncol 2024; 14:1411432. [PMID: 39534096 PMCID: PMC11554526 DOI: 10.3389/fonc.2024.1411432] [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: 04/02/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024] Open
Abstract
Lung cancer is the second most common type of cancer and is the leading cause of cancer-related deaths in the United States. Approximately 10-40% of patients with solid tumors develop brain metastases, with non-small cell lung cancer accounting for approximately 50% of all cases of patients with brain metastases. Many management options are available which can include surgery, radiation, and systemic therapy. A variety of factors go into the selection of management of brain metastases. In this review, we will focus on the treatment strategies and optimizing the management of brain metastases in patients with non-small cell lung cancer.
Collapse
Affiliation(s)
- Parth J. Sampat
- Division of Hematology and Medical Oncology, Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Alyssa Cortese
- Division of Hematology and Medical Oncology, Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Alexandra Goodman
- Division of Hematology and Medical Oncology, Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Ghanshyam H. Ghelani
- Division of Hematology and Medical Oncology, Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Michael D. Mix
- Department of Radiation Oncology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Stephen Graziano
- Division of Hematology and Medical Oncology, Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Alina Basnet
- Division of Hematology and Medical Oncology, Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
45
|
Jayakrishnan R, Kwiatkowski DJ, Rose MG, Nassar AH. Topography of mutational signatures in non-small cell lung cancer: emerging concepts, clinical applications, and limitations. Oncologist 2024; 29:833-841. [PMID: 38907669 PMCID: PMC11449018 DOI: 10.1093/oncolo/oyae091] [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: 12/08/2023] [Accepted: 04/16/2024] [Indexed: 06/24/2024] Open
Abstract
The genome of a cell is continuously battered by a plethora of exogenous and endogenous processes that can lead to damaged DNA. Repair mechanisms correct this damage most of the time, but failure to do so leaves mutations. Mutations do not occur in random manner, but rather typically follow a more or less specific pattern due to known or imputed mutational processes. Mutational signature analysis is the process by which the predominant mutational process can be inferred for a cancer and can be used in several contexts to study both the genesis of cancer and its response to therapy. Recent pan-cancer genomic efforts such as "The Cancer Genome Atlas" have identified numerous mutational signatures that can be categorized into single base substitutions, doublet base substitutions, or small insertions/deletions. Understanding these mutational signatures as they occur in non-small lung cancer could improve efforts at prevention, predict treatment response to personalized treatments, and guide the development of therapies targeting tumor evolution. For non-small cell lung cancer, several mutational signatures have been identified that correlate with exposures such as tobacco smoking and radon and can also reflect endogenous processes such as aging, APOBEC activity, and loss of mismatch repair. Herein, we provide an overview of the current knowledge of mutational signatures in non-small lung cancer.
Collapse
Affiliation(s)
- Ritujith Jayakrishnan
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - David J Kwiatkowski
- Department of Pulmonary Medicine, Brigham and Women's Hospital, Boston, MA, 02115, United States
| | - Michal G Rose
- Yale University School of Medicine and Cancer Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, United States
- Department of Medicine, Medical Oncology Division, Yale Cancer Center, New Haven, CT, United States
| | - Amin H Nassar
- Yale University School of Medicine and Cancer Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, United States
| |
Collapse
|
46
|
Gupta B, Gosa Barrett L, Liu SV. NRG1 Fusions in NSCLC: Being eNRGy Conscious. LUNG CANCER (AUCKLAND, N.Z.) 2024; 15:143-148. [PMID: 39376790 PMCID: PMC11457762 DOI: 10.2147/lctt.s464626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 08/27/2024] [Indexed: 10/09/2024]
Abstract
Fusions in neuregulin 1 (NRG1) are rare oncogenic drivers that occur across a number of tumor types, including non-small cell lung cancer (NSCLC). NRG1 has an EGF-like domain that serves as a ligand for HER3 receptors, inducing heterodimerization, usually with HER2, and subsequent activation of oncogenic downstream signaling pathways. Emerging evidence suggests that NSCLC harboring NRG1 fusions do not respond as well to standard therapeutic options including chemotherapy and immunotherapy, and prognosis is poor. Novel treatment approaches targeting the HER2/HER3 pathway are under investigation. Here, we discuss the biology and detection of NRG1 fusions in NSCLC and promising targeted treatment strategies for tumors harboring the mutation.
Collapse
Affiliation(s)
- Brinda Gupta
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Laura Gosa Barrett
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Stephen V Liu
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| |
Collapse
|
47
|
Vitale A, Mastrantoni L, Russo J, Giacomini F, Giannarelli D, Duranti S, Vita E, Nero C, D'Argento E, Pasciuto T, Giacò L, Di Salvatore M, Panfili A, Stefani A, Cancellieri A, Lococo F, De Paolis E, Livi V, Daniele G, Trisolini R, Minucci A, Margaritora S, Lorusso D, Normanno N, Scambia G, Tortora G, Bria E. Impact of Comprehensive Genome Profiling on the Management of Advanced Non-Small Cell Lung Cancer: Preliminary Results From the Lung Cancer Cohort of the FPG500 Program. JCO Precis Oncol 2024; 8:e2400297. [PMID: 39374480 DOI: 10.1200/po.24.00297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/02/2024] [Accepted: 08/16/2024] [Indexed: 10/09/2024] Open
Abstract
PURPOSE The clinical and research FPG500 program (ClinicalTrials.gov identifier: NCT06020625) is currently ongoing at the Fondazione Policlinico Universitario Agostino Gemelli IRCCS to tailor matched targeted therapies (MTTs) according to biomarkers predictive of response identified by comprehensive genome profiling (CGP). MATERIALS AND METHODS The non-small cell lung cancer (NSCLC) cohort results from the FPG500 program are outlined. CGP was performed by TruSight Oncology 500 High Throughput (TSO500HT) assay or Oncomine Focus Assay plus Archer's FusionPlex Lung Panel according to tumor cell content and DNA/RNA quantity. Relevant issues for Molecular Tumor Board (MTB) evaluation included uncommon genomic findings, evaluation for off-label therapies, uncertain result confirmation, and variants of suspect germline origin requiring genetic counseling. Progression-free survival (PFS) and overall survival (OS) for the enrolled patients were assessed using Kaplan-Meier analysis. RESULTS In 2022, 283 patients with NSCLC were considered for sequencing, with 93% meeting eligibility criteria. TSO500HT sequencing was conducted in 76% of patients. Follow-up data were obtained for 187 patients, among whom 81% received treatment. Potential driver alterations were identified in 59% of patients, with 41% receiving MTT: 25% were prescribed approved MTTs, whereas 16% gained access to experimental drugs post-MTB evaluation; of note, 18% did not receive any MTT because the regimen was not yet reimbursed in our country. Median PFS and OS varied among treatment groups, with standard chemotherapy/immunotherapy at 7.7 and 10.7 months, approved tyrosine kinase inhibitors at 18.8 and 23.9 months, and MTT post-MTB discussion at 14 and 23.4 months, respectively. CONCLUSION The early data of the FPG program (NSCLC cohort) support the implementation of CGP and MTB in clinical practice to grant access to patients harboring actionable molecular alterations to the most effective and individualized available treatment options, thus improving their survival outcomes.
Collapse
Affiliation(s)
- Antonio Vitale
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Mastrantoni
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Jacopo Russo
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Flavia Giacomini
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Scientific Directorate, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Diana Giannarelli
- Biostatistical Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Simona Duranti
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Scientific Directorate, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Emanuele Vita
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Camilla Nero
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Women, Children and Public Health Sciences Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ettore D'Argento
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Tina Pasciuto
- Data Collection Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luciano Giacò
- Bioinformatics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Mariantonietta Di Salvatore
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Arianna Panfili
- Scientific Directorate, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Alessio Stefani
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandra Cancellieri
- Pathology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Filippo Lococo
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Thoracic Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Elisa De Paolis
- Laboratory of Clinical Molecular and Personalized Diagnostics, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Vanina Livi
- Interventional Pulmonology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gennaro Daniele
- Phase 1 Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Rocco Trisolini
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Interventional Pulmonology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Angelo Minucci
- Laboratory of Clinical Molecular and Personalized Diagnostics, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Stefano Margaritora
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Thoracic Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Domenica Lorusso
- Gynecologic Oncology Unit, Humanitas San Pio X, Humanitas University, Milan, Italy
| | - Nicola Normanno
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori," Meldola, Italy
| | - Giovanni Scambia
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Women, Children and Public Health Sciences Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giampaolo Tortora
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Medical Oncology, Ospedale Isola Tiberina-Gemelli Isola, Rome, Italy
| |
Collapse
|
48
|
Ha H, Lee HY, Kim JH, Kim DY, An HJ, Bae S, Park HS, Kang JH. Precision Oncology Clinical Trials: A Systematic Review of Phase II Clinical Trials with Biomarker-Driven, Adaptive Design. Cancer Res Treat 2024; 56:991-1013. [PMID: 38726510 PMCID: PMC11491240 DOI: 10.4143/crt.2024.128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/29/2024] [Indexed: 08/30/2024] Open
Abstract
Novel clinical trial designs are conducted in the precision medicine era. This study aimed to evaluate biomarker-driven, adaptive phase II trials in precision oncology, focusing on infrastructure, efficacy, and safety. We systematically reviewed and analyzed the target studies. EMBASE and PubMed searches from 2015 to 2023 generated 29 eligible trials. Data extraction included infrastructure, biomarker screening methodologies, efficacy, and safety profiles. Government agencies, cancer hospitals, and academic societies with accumulated experiences led investigator-initiated precision oncology clinical trials (IIPOCTs), which later guided sponsor-initiated precision oncology clinical trials (SIPOCTs). Most SIPOCTs were international studies with basket design. IIPOCTs primarily used the central laboratory for biomarker screening, but SIPOCTs used both central and local laboratories. Most of the studies adapted next-generation sequencing and/or immunohistochemistry for biomarker screening. Fifteen studies included an independent central review committee for outcome investigation. Efficacy assessments predominantly featured objective response rate as the primary endpoint, with varying results. Nine eligible studies contributed to the United States Food and Drug Administration's marketing authorization. Safety monitoring was rigorous, but reporting formats lacked uniformity. Health-related quality of life and patient-reported outcomes were described in some protocols but rarely reported. Our results reveal that precision oncology trials with adaptive design rapidly and efficiently evaluate anticancer drugs' efficacy and safety, particularly in specified biomarker-driven cohorts. The evolution from IIPOCT to SIPOCT has facilitated fast regulatory approval, providing valuable insights into the precision oncology landscape.
Collapse
Affiliation(s)
- Hyerim Ha
- Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - Hee Yeon Lee
- Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Do Yeun Kim
- Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Ho Jung An
- Division of Oncology, Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - SeungJin Bae
- College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Hye-sung Park
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jin Hyoung Kang
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
49
|
Okuno T, Isobe T, Tsubata Y. Current pharmacologic treatment of brain metastasis in non-small cell lung cancer. Clin Exp Metastasis 2024; 41:549-565. [PMID: 38466521 PMCID: PMC11499348 DOI: 10.1007/s10585-024-10276-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/28/2024] [Indexed: 03/13/2024]
Abstract
Lung cancer is a type of cancer that can metastasize to the lungs, brain, bones, liver, adrenal glands, and other organs; however, the occurrence of brain metastases is the most common event. Symptoms of brain metastasis include motor dysfunction, mental dysfunction, seizures, headaches, nausea, and vomiting, and significantly reduce the quality of life of cancer patients. Brain metastases are a poor prognostic factor, and controlling them is extremely important for prolonging prognosis and improving the quality of life. Currently, local surgery and radiotherapy are recommended for their treatment. However, recently, cancer treatments using molecular-targeted drugs and immune checkpoint inhibitors have been introduced, which may also be effective against brain metastases. Therefore, it is necessary to determine whether local or systemic therapy is optimal for each case. In this review, we focus on recent findings regarding drug therapy in treating brain metastases from advanced non-small cell lung cancer.
Collapse
Affiliation(s)
- Takae Okuno
- Division of Medical Oncology and Respiratory Medicine, Department of Internal Medicine, Shimane University Faculty of Medicine, 89-1, Enyacho, Izumo, Shimane, 693-8501, Japan
| | - Takeshi Isobe
- Division of Medical Oncology and Respiratory Medicine, Department of Internal Medicine, Shimane University Faculty of Medicine, 89-1, Enyacho, Izumo, Shimane, 693-8501, Japan
| | - Yukari Tsubata
- Division of Medical Oncology and Respiratory Medicine, Department of Internal Medicine, Shimane University Faculty of Medicine, 89-1, Enyacho, Izumo, Shimane, 693-8501, Japan.
| |
Collapse
|
50
|
Chunmao W, Haijie C, Zitong W, Zhi Y. A case of neoadjuvant targeted therapy with pralsetinib for locally advanced lung adenocarcinoma with RET fusion mutation. J Cardiothorac Surg 2024; 19:554. [PMID: 39354540 PMCID: PMC11443678 DOI: 10.1186/s13019-024-03092-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 09/15/2024] [Indexed: 10/03/2024] Open
Abstract
This case report details the successful treatment of a 68-year-old male patient with locally advanced RET-rearranged lung adenocarcinoma using neoadjuvant pralsetinib. The patient initially presented with a suspicious right upper lobe nodule, which was later diagnosed as lung adenocarcinoma following genetic testing that revealed a RET exon 12 fusion. After 2 months of neoadjuvant treatment with pralsetinib, a significant radiological response was observed, with a reduction in tumor size and metabolic activity. Subsequently, the patient underwent video-assisted thoracoscopic right upper lobectomy and mediastinal lymph node dissection. Postoperative pathological analysis revealed a major pathological response, with only 5% residual tumor cells in the primary lesion and no viable tumor cells in the lymph nodes. Postoperative pathological staging of TNM was ypT1aN0M0, stage IA1(AJCC, 8th edition). The patient recovered well after surgery, demonstrating the potential efficacy of neoadjuvant pralsetinib in locally advanced RET-rearranged lung adenocarcinoma. However, further clinical validation is required to establish the role of neoadjuvant targeted therapy and postoperative adjuvant therapy in this patient population.
Collapse
Affiliation(s)
- Wang Chunmao
- Department of Thoracic Surgery, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, No. 9 Beiguan Street, Tongzhou District, Beijing, 101149, China
| | - Cheng Haijie
- Department of Thoracic Surgery, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, No. 9 Beiguan Street, Tongzhou District, Beijing, 101149, China
| | - Wang Zitong
- Department of Thoracic Surgery, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, No. 9 Beiguan Street, Tongzhou District, Beijing, 101149, China
| | - Yang Zhi
- Department of Thoracic Surgery, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, No. 9 Beiguan Street, Tongzhou District, Beijing, 101149, China.
| |
Collapse
|