1
|
Zhu S, Fu K, Li S, Yang C, Pan C, Wang X, Wang F, Yu X, To KKW, Fu L. Cardiotoxicity of small-molecule kinase inhibitors in cancer therapy. Exp Hematol Oncol 2025; 14:68. [PMID: 40346640 PMCID: PMC12063284 DOI: 10.1186/s40164-025-00660-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Cancer is one of the leading causes of death worldwide. Recent advances in precision oncology have enabled many specific cancer patient populations to respond well and achieve longer survival with small-molecule kinase inhibitors, which have become a new therapeutic strategy for tumors. Since 2001, the Food and Drug Administration has approved 108 and 63 new anticancer drugs for treating solid tumors and hematological malignancies, respectively, 89 of which belong to the large group of small-molecule kinase inhibitors (SMKIs). Compared to conventional chemotherapeutic agents such as cyclophosphamide, doxorubicin, and 5-FU, SMKIs offer better efficacy with fewer toxic side effects. Nevertheless, with the development of more novel SMKIs and their wider clinical application to a larger population of cancer patients, variable degrees of cardiotoxic adverse events have emerged for some SMKIs during cancer therapy. This review comprehensively summarizes the most updated progress in the cardiotoxicity of SMKIs in cancer therapy and discusses the new findings and mechanisms, which will provide emerging strategies for the prevention of cardiotoxicity caused by small molecule targeted drugs and the design of the next generation of low cardiotoxicity targeted drugs.
Collapse
Affiliation(s)
- Shuangli Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Kai Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Sijia Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Can Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xueping Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xiyong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangdong, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
2
|
Maleddu A, Hinz TK, Black MA, Aisner DL, Marshall CB, Elias AD, Wilky BA, Heasley LE, Davies KD. Novel PLEC-EML4-ALK Double Fusion Underlying Crizotinib Resistance in a Metastatic Inflammatory Myofibroblastic Tumor: A Case Report. JTO Clin Res Rep 2025; 6:100791. [PMID: 40225953 PMCID: PMC11986212 DOI: 10.1016/j.jtocrr.2025.100791] [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: 09/06/2024] [Revised: 11/25/2024] [Accepted: 12/31/2024] [Indexed: 04/15/2025] Open
Abstract
ALK fusions are frequent oncogenic drivers in inflammatory myofibroblastic tumors. Treatment with crizotinib is effective in fusion-positive patients; however, acquired resistance remains a challenge. Here, we present a case of EML4-ALK-positive metastatic inflammatory myofibroblastic tumor that initially responded to crizotinib but developed resistance. The progressing lesion revealed the acquisition of a "double fusion" event in which EML4-ALK was additionally fused to PLEC to create a PLEC-EML4-ALK transcript. The double fusion was associated with an increase in ALK expression, mimicking the ALK fusion amplification that is a known mechanism of resistance to crizotinib in lung cancer. On transition to the more potent ALK inhibitor alectinib, the patient exhibited a dramatic response. Thus, the formation of a double fusion represents a novel and targetable mechanism of resistance to crizotinib.
Collapse
Affiliation(s)
- Alessandra Maleddu
- Department of Medicine, University of Colorado – Anschutz Medical Campus, Aurora, Colorado
| | - Trista K. Hinz
- Department of Craniofacial Biology, University of Colorado – Anschutz Medical Campus, Aurora, Colorado
| | - Margaret A. Black
- Department of Pathology, University of Colorado – Anschutz Medical Campus, Aurora, Colorado
| | - Dara L. Aisner
- Department of Pathology, University of Colorado – Anschutz Medical Campus, Aurora, Colorado
| | - Carrie B. Marshall
- Department of Pathology, University of Colorado – Anschutz Medical Campus, Aurora, Colorado
| | - Anthony D. Elias
- Department of Medicine, University of Colorado – Anschutz Medical Campus, Aurora, Colorado
| | - Breelyn A. Wilky
- Department of Medicine, University of Colorado – Anschutz Medical Campus, Aurora, Colorado
| | - Lynn E. Heasley
- Department of Craniofacial Biology, University of Colorado – Anschutz Medical Campus, Aurora, Colorado
| | - Kurtis D. Davies
- Department of Pathology, University of Colorado – Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
3
|
Voena C, Ambrogio C, Iannelli F, Chiarle R. ALK in cancer: from function to therapeutic targeting. Nat Rev Cancer 2025; 25:359-378. [PMID: 40055571 DOI: 10.1038/s41568-025-00797-9] [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] [Accepted: 02/04/2025] [Indexed: 05/01/2025]
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase (RTK) that acts as an oncogenic driver in solid and haematological malignancies in both children and adults. Although ALK-expressing (ALK+) tumours show strong initial responses to the series of ALK inhibitors currently available, many patients will develop resistance. In this Review, we discuss recent advances in ALK oncogenic signalling, together with existing and promising new modalities to treat ALK-driven tumours, including currently approved ALK-directed therapies, namely tyrosine kinase inhibitors, and novel approaches such as ALK-specific immune therapies. Although ALK inhibitors have changed the management and clinical history of ALK+ tumours, they are still insufficient to cure most of the patients. Therefore, more effort is needed to further improve outcomes and prevent the tumour resistance, recurrence and metastatic spread that many patients with ALK+ tumours experience. Here, we outline how a multipronged approach directed against ALK and other essential pathways that sustain the persistence of ALK+ tumours, together with potent or specific immunotherapies, could achieve this goal. We envision that the lessons learned from treating ALK+ tumours in the clinic could ultimately accelerate the implementation of innovative combination therapies to treat tumours driven by other tyrosine kinases or oncogenes with similar properties.
Collapse
Affiliation(s)
- Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Fabio Iannelli
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy.
- Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Zha Z, Liu C, Yan M, Chen C, Yu C, Chen Y, Zhou C, Li L, Li YC, Yamaguchi H, Ye L, Liu T, Wang YN, Lee HH, Yang WH, Chan LC, Ke B, Hsu JL, Ding L, Ji D, Pan P, Meng Y, Pu Y, Liu L, Hung MC. RNase1-driven ALK-activation is an oncogenic driver and therapeutic target in non-small cell lung cancer. Signal Transduct Target Ther 2025; 10:124. [PMID: 40246819 PMCID: PMC12006399 DOI: 10.1038/s41392-025-02206-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/18/2025] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Targeted therapy has achieved significant success in the treatment of non-small cell lung cancer (NSCLC), particularly in patients harboring common oncogenic driver mutations such as EGFR, KRAS, and ALK rearrangement. However, ~35-50% of NSCLC patients without tyrosine kinase mutation or rearrangement (non-mutated) cannot benefit from these targeted treatments, highlighting the urgent need for novel therapeutic strategies for this patient population. In this study, we report a non-canonical role of human secretory ribonuclease 1 (RNase1), which binds to and activates wild-type ALK in lung cancer cells, thereby triggering its downstream signaling pathway. RNase1-driven ALK-activation (RDAA) cells exhibit enhanced cell proliferation, migration, and colony formation. Additionally, RDAA facilitates tumor formation in fibroblast models, further underscoring its oncogenic potential in vivo. Importantly, RDAA lung cancer cells exhibit marked sensitivity to FDA-approved ALK inhibitors. Tumor growth suppression and survival were substantially improved in both RDAA-positive NSCLC cell line-derived and patient-derived xenograft tumor models treated with ALK inhibitors. Monoclonal antibodies against RNase1 and phosphorylated-ALK were used to analyze two different human NSCLC tissue cohorts by immunohistochemical staining identified 10.4% (5/48) and 8.5% (100/1173) patients who were RDAA positive, respectively. Notably, among the nine RDAA-positive NSCLC patients who accepted ALK inhibitor treatment, five achieved objective response including two who experienced complete response (CR). Together, the current study identifies RDAA as an oncogenic driver and proposes an effective targeted therapy strategy for non-mutated NSCLC patients.
Collapse
Affiliation(s)
- Zhengyu Zha
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, University of Sichuan, Chengdu, Sichuan, China
| | - Chunxiao Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Meisi Yan
- Department of Pathology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Cong Chen
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, University of Sichuan, Chengdu, Sichuan, China
| | - Cheng Yu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, University of Sichuan, Chengdu, Sichuan, China
| | - Yaohui Chen
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, University of Sichuan, Chengdu, Sichuan, China
| | - Chenhao Zhou
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Liver Cancer and Transplant, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Lu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi-Chuan Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hiro Yamaguchi
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Leiguang Ye
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tong Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying-Nai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heng-Huan Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wen-Hao Yang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li-Chuan Chan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Baozhen Ke
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer L Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Dong Ji
- Betta Pharmaceuticals Co. Ltd, Hangzhou, China
| | - Peng Pan
- Betta Pharmaceuticals Co. Ltd, Hangzhou, China
| | - Yiran Meng
- Hangzhou Repugene Technology Co., Ltd, Hangzhou, China
| | - Yue Pu
- Hangzhou Repugene Technology Co., Ltd, Hangzhou, China
| | - Lunxu Liu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, University of Sichuan, Chengdu, Sichuan, China.
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
5
|
Choi JH. Inflammatory Myofibroblastic Tumor: An Updated Review. Cancers (Basel) 2025; 17:1327. [PMID: 40282503 PMCID: PMC12026078 DOI: 10.3390/cancers17081327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/12/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025] Open
Abstract
Inflammatory myofibroblastic tumor (IMT) is a rare neoplasm that is characterized by a proliferation of myofibroblastic and fibroblastic spindle cells, accompanied by an inflammatory infiltrate that is abundant in plasma cells, lymphocytes, and eosinophils. IMT can arise in various anatomical locations but most commonly occurs in the abdominal cavity, retroperitoneum, and lung, particularly in children and young adults. IMT typically demonstrates local invasion or recurrence, whereas metastasis is rare. IMTs pose a diagnostic challenge because of their overlapping morphological characteristics with a heterogeneous group of nonneoplastic and neoplastic lesions. Precise diagnosis is crucial for optimal management and accurate prognostication. Despite recent advancements in IMT diagnosis and treatment, its biological complexity and clinical management remain challenging due to significant histological heterogeneity and molecular genetic diversity. This review provides comprehensive updates on the clinical, molecular, and pathological characteristics of IMT, highlighting the diagnostic approaches and key differential diagnoses.
Collapse
Affiliation(s)
- Joon Hyuk Choi
- Department of Pathology, Yeungnam University College of Medicine, Daegu 42415, Republic of Korea
| |
Collapse
|
6
|
Park S, Haam K, Heo H, Kim D, Kim M, Jung H, Cha S, Kim M, Lee H. Integrative transcriptomic analysis identifies emetine as a promising candidate for overcoming acquired resistance to ALK inhibitors in lung cancer. Mol Oncol 2025; 19:1155-1169. [PMID: 39540457 PMCID: PMC11977641 DOI: 10.1002/1878-0261.13738] [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: 02/16/2024] [Revised: 08/02/2024] [Accepted: 09/10/2024] [Indexed: 11/16/2024] Open
Abstract
Anaplastic lymphoma kinase (ALK; also known as ALK tyrosine kinase receptor) inhibitors (ALKi) are effective in treating lung cancer patients with chromosomal rearrangement of ALK. However, continuous treatment with ALKis invariably leads to acquired resistance in cancer cells. In this study, we propose an efficient strategy to suppress ALKi resistance through a meta-analysis of transcriptome data from various cell models of acquired resistance to ALKis. We systematically identified gene signatures that consistently showed altered expression during the development of resistance and conducted computational drug screening using these signatures. We identified emetine as a promising candidate compound to inhibit the growth of ALKi-resistant cells. We demonstrated that emetine exhibited effectiveness in inhibiting the growth of ALKi-resistant cells, and further interpreted its impact on the resistant signatures through drug-induced RNA-sequencing data. Our transcriptome-guided systematic approach paves the way for efficient drug discovery to overcome acquired resistance to cancer therapy.
Collapse
Affiliation(s)
- Sang‐Min Park
- College of PharmacyChungnam National UniversityDaejeonKorea
| | - Keeok Haam
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
| | - Haejeong Heo
- Personalized Genomic Medicine Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
- Department of Functional GenomicsUniversity of Science and Technology (UST)DaejeonKorea
| | - Doyeong Kim
- College of PharmacyChungnam National UniversityDaejeonKorea
| | - Min‐Ju Kim
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug DevelopmentPusan National UniversityBusanKorea
| | - Hyo‐Jung Jung
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
| | - Seongwon Cha
- Korean Medicine (KM) Data DivisionKorea Institute of Oriental MedicineDaejeonKorea
| | - Mirang Kim
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
- Personalized Genomic Medicine Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
- Department of Functional GenomicsUniversity of Science and Technology (UST)DaejeonKorea
| | - Haeseung Lee
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug DevelopmentPusan National UniversityBusanKorea
| |
Collapse
|
7
|
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
|
8
|
Morand S, Rager L, Craig D, Nemunaitis A, Choucair K, Rao D, Stanbery L, Phinney RC, Walter A, Ghisoli M, Nemunaitis J. Clinical characterization and therapeutic targeting of fusion genes in oncology. Future Oncol 2025; 21:1249-1260. [PMID: 40128124 PMCID: PMC11988278 DOI: 10.1080/14796694.2025.2477974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Gene fusions represent important oncogenic driver mutations resulting in aberrant cellular signaling. In up to 17% of all solid tumors at least one gene fusion can be identified. Precision therapy targeting fusion gene signaling has demonstrated effective clinical benefit. Advancements in clinically relevant next-generation sequencing and bioinformatic techniques have enabled expansion of therapeutic opportunity to subpopulations of patients with fusion gene expression. Clinically, tyrosine inhibitors have shown efficacy in treating fusion gene expressing cancers. Fusion genes are also clonal mutations, meaning it is a personal cancer target involving all cancer cells of that patient, not just a subpopulation of cancer cells within the cancer mass. Thus, both fusion signal disruption and immune signal targeting are effective therapeutic directions. This review discusses fusion gene targeting, therapeutic resistance, and molecular biomarkers.
Collapse
Affiliation(s)
- Susan Morand
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Lauren Rager
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Daniel Craig
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | | | - Khalil Choucair
- Department of Hematology/Oncology, Barbara Karmanos Cancer Institute,Wayne State University, Detroit, MI, USA
| | - Donald Rao
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
| | - Laura Stanbery
- Taylor Cancer Research Center, Maumee, OH, USA
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
| | - Richard C. Phinney
- Taylor Cancer Research Center, Maumee, OH, USA
- Department of Hematology/Oncology, Toledo Clinic Cancer Center, Maumee, OH, USA
| | - Adam Walter
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
- Department of Gynecologic Oncology, Promedica Health System, Toledo, OH, USA
| | - Maurizio Ghisoli
- Department of Pediatric Hematology/Oncology, Texas Oncology, P.A, Dallas, TX, USA
| | - John Nemunaitis
- Taylor Cancer Research Center, Maumee, OH, USA
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
- Department of Hematology/Oncology, Toledo Clinic Cancer Center, Maumee, OH, USA
| |
Collapse
|
9
|
Schneider JL, Kurmi K, Dai Y, Dhiman I, Joshi S, Gassaway BM, Johnson CW, Jones N, Li Z, Joschko CP, Fujino T, Paulo JA, Yoda S, Baquer G, Ruiz D, Stopka SA, Kelley L, Do A, Mino-Kenudson M, Sequist LV, Lin JJ, Agar NYR, Gygi SP, Haigis KM, Hata AN, Haigis MC. GUK1 activation is a metabolic liability in lung cancer. Cell 2025; 188:1248-1264.e23. [PMID: 39919745 PMCID: PMC12148050 DOI: 10.1016/j.cell.2025.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/14/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025]
Abstract
Little is known about metabolic vulnerabilities in oncogene-driven lung cancer. Here, we perform a phosphoproteomic screen in anaplastic lymphoma kinase (ALK)-rearranged ("ALK+") patient-derived cell lines and identify guanylate kinase 1 (GUK1), a guanosine diphosphate (GDP)-synthesizing enzyme, as a target of ALK signaling in lung cancer. We demonstrate that ALK binds to and phosphorylates GUK1 at tyrosine 74 (Y74), resulting in increased GDP biosynthesis. Spatial imaging of ALK+ patient tumor specimens shows enhanced phosphorylation of GUK1 that significantly correlates with guanine nucleotides in situ. Abrogation of GUK1 phosphorylation reduces intracellular GDP and guanosine triphosphate (GTP) pools and decreases mitogen-activated protein kinase (MAPK) signaling and Ras-GTP loading. A GUK1 variant that cannot be phosphorylated (Y74F) decreases tumor proliferation in vitro and in vivo. Beyond ALK, other oncogenic fusion proteins in lung cancer also regulate GUK1 phosphorylation. These studies may pave the way for the development of new therapeutic approaches by exploiting metabolic dependencies in oncogene-driven lung cancers.
Collapse
Affiliation(s)
- Jaime L Schneider
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Kiran Kurmi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Yutong Dai
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Department of Chemistry and Chemical Biology, Harvard University, Boston, MA, USA
| | - Ishita Dhiman
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Shakchhi Joshi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Brandon M Gassaway
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | | | - Nicole Jones
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Zongyu Li
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Christian P Joschko
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Toshio Fujino
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Joao A Paulo
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Satoshi Yoda
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Gerard Baquer
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniela Ruiz
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sylwia A Stopka
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Liam Kelley
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew Do
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lecia V Sequist
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jessica J Lin
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Kevin M Haigis
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Aaron N Hata
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Zhang W, Qi L, Xu H, Yin C, Yu Z, Xu R, Feng C, Ren X, Tu C, Li Z. Cooperative blockade of FLT3 and ALK synergistically suppresses growth of osteosarcoma. Oncogene 2025; 44:427-438. [PMID: 39562656 DOI: 10.1038/s41388-024-03205-y] [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: 06/09/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024]
Abstract
Osteosarcoma is a common primary malignant bone tumor in children and young adults, with limited progress in improving survival rates for metastatic or recurrent cases. Kinase inhibitors have emerged as potential treatments for osteosarcoma due to the critical role kinases play in regulating cellular networks. However, single-agent kinase inhibitors often face challenges due to the activation of compensatory oncogenic signaling pathways, which can undermine treatment efficacy. In this study, a combination screening of FDA-approved kinase inhibitors was conducted in osteosarcoma cells. We identified the combination of ALK inhibitor and FLT3 inhibitor as a potent kinase-based therapeutic strategy for osteosarcoma. Our results showed that the combinatorial treatment synergistically suppressed osteosarcoma in cell lines, patient-derived organoids, and xenograft models. Mechanistically, the inhibition of FLT3 significantly promoted the activation of ALK, which subsequently enhanced its downstream PI3K/Akt and MAPK signaling pathways. The combinatorial use of an ALK inhibitor could reverse this process. Thus, our study demonstrates that the cooperative blockade of FLT3 and ALK synergistically suppresses osteosarcoma, providing a potential alternative for its treatment.
Collapse
Affiliation(s)
- Wenchao Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Haodong Xu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Chi Yin
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Zhuowen Yu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Ruiling Xu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Chengyao Feng
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Xiaolei Ren
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China.
| | - Chao Tu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China.
- Changsha Medical University, Changsha, China.
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China.
- Shenzhen Research Institute of Central South University, Guangdong, China.
- FuRong Laboratory, Changsha, Hunan, China.
| |
Collapse
|
11
|
Waliany S, Lin JJ, Gainor JF. Evolution of first versus next-line targeted therapies for metastatic non-small cell lung cancer. Trends Cancer 2025; 11:245-257. [PMID: 39890507 DOI: 10.1016/j.trecan.2025.01.005] [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: 09/25/2024] [Revised: 12/21/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Abstract
The expanding armamentarium of targeted therapies has revolutionized treatment for metastatic oncogene-addicted lung cancers. For multiple subsets, such as those harboring EGFR mutations and fusions in ALK or ROS1, successive generation of increasingly potent, selective, and brain-penetrating targeted therapies have shifted the treatment paradigm towards preferential first-line use of next-generation drugs. This evolution in clinical practice provides a lens through which to review the lessons learned from drug development in oncogene-addicted lung cancers, guided by translational insights into tumor biology and mechanisms of therapeutic resistance. For oncogenic drivers that are less sensitive to single-agent targeted therapies, rationally designed combination strategies will be needed to enable first-line use of targeted agents.
Collapse
Affiliation(s)
- Sarah Waliany
- Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica J Lin
- Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Justin F Gainor
- Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
12
|
Sikkema BJ, Baart SJ, Paats MS, Smit EF, Schols AMWJ, Mathijssen RHJ, van Rossum EFC, Dingemans AMC. Body Weight Gain Associated With Alectinib in Patients With ALK+ Non-Small Cell Lung Cancer: Pooled Analysis of Individual Patient Data From Four Prospective Clinical Trials. J Clin Oncol 2025; 43:641-650. [PMID: 39661917 DOI: 10.1200/jco-24-01579] [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: 07/19/2024] [Revised: 09/30/2024] [Accepted: 10/31/2024] [Indexed: 12/13/2024] Open
Abstract
PURPOSE Weight gain is a known adverse event (AE) of alectinib. This study evaluates the progression of actual weight gain over time and explores its association with baseline characteristics. METHODS A pooled analysis of individual patient data from four clinical trials (ALEX, J-ALEX, ALUR, and ML29453) was conducted. Actual weight gain was calculated as the percent change from baseline. A linear mixed model estimated weight change over time and associations between clinical characteristics and weight change. RESULTS Follow-up weights were available for three trials (J-ALEX, ALUR, and ML29453) and missing for ALEX. In total, 2,622 weights were recorded in the first year (N = 302). At baseline, 13.6% of the Japanese population were underweight and 5.0% in the Western population. Actual weight gain of any grade was substantially higher than reported AE rates (49% v 5%), with 18% experiencing ≥10% weight gain (from median 55.6 kg to 64.1 kg). Time on alectinib was positively associated with weight change (β = .37; 95% CI, 0.24 to 0.51; P < .001), corresponding to an average increase of 4.4% over 1 year. Baseline BMI was not associated with weight change in J-ALEX (β = -.090 [95% CI, -0.19 to 0.012]; P = .092) and ALUR/ML29453 (β = -.016 [95% CI, -0.077 to 0.044]; P = .59). Baseline albumin was positively associated with weight change in ALUR/ML29453 (β = .084 [95% CI, 0.027 to 0.14]; P = .0045), although not considered a clinically meaningful predictor. CONCLUSION Weight gain is under-reported as AE in trials. Actual weights showed ≥10% weight gain in 18% of patients. Clinicians should be aware of this AE, emphasizing the importance of timely identification and monitoring weight. Identifying predictors for weight gain remains challenging.
Collapse
Affiliation(s)
- Barend J Sikkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sara J Baart
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Biostatistics, Erasmus MC, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marthe S Paats
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Egbert F Smit
- Department of Pulmonary Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Annemie M W J Schols
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Elisabeth F C van Rossum
- Department of Internal Medicine, Division of Endocrinology and Obesity Center CGG, Erasmus MC, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Anne-Marie C Dingemans
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
13
|
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
|
14
|
De Lucia A, Mazzotti L, Gaimari A, Zurlo M, Maltoni R, Cerchione C, Bravaccini S, Delmonte A, Crinò L, Borges de Souza P, Pasini L, Nicolini F, Bianchi F, Juan M, Calderon H, Magnoni C, Gazzola L, Ulivi P, Mazza M. Non-small cell lung cancer and the tumor microenvironment: making headway from targeted therapies to advanced immunotherapy. Front Immunol 2025; 16:1515748. [PMID: 39995659 PMCID: PMC11847692 DOI: 10.3389/fimmu.2025.1515748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
Over the past decades, significant progress has been made in the understanding of non-small cell lung cancer (NSCLC) biology and tumor progression mechanisms, resulting in the development of novel strategies for early detection and wide-ranging care approaches. Since their introduction, over 20 years ago, targeted therapies with tyrosine kinase inhibitors (TKIs) have revolutionized the treatment landscape for NSCLC. Nowadays, targeted therapies remain the gold standard for many patients, but still they suffer from many adverse effects, including unexpected toxicity and intrinsic acquired resistance mutations, which lead to relapse. The adoption of immune checkpoint inhibitors (ICIs) in 2015, has offered exceptional survival benefits for patients without targetable alterations. Despite this notable progress, challenges remain, as not all patients respond favorably to ICIs, and resistance to therapy can develop over time. A crucial factor influencing clinical response to immunotherapy is the tumor microenvironment (TME). The TME is pivotal in orchestrating the interactions between neoplastic cells and the immune system, influencing tumor growth and treatment outcomes. In this review, we discuss how the understanding of this intricate relationship is crucial for the success of immunotherapy and survey the current state of immunotherapy intervention, with a focus on forthcoming and promising chimeric antigen receptor (CAR) T cell therapies in NSCLC. The TME sets major obstacles for CAR-T therapies, creating conditions that suppress the immune response, inducing T cell exhaustion. To enhance treatment efficacy, specific efforts associated with CAR-T cell therapy in NSCLC, should definitely focus TME-related immunosuppression and antigen escape mechanisms, by combining CAR-T cells with immune checkpoint blockades.
Collapse
Affiliation(s)
- Anna De Lucia
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucia Mazzotti
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Anna Gaimari
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Matteo Zurlo
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Roberta Maltoni
- Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sara Bravaccini
- Department of Medicine and Surgery, “Kore” University of Enna, Enna, Italy
| | - Angelo Delmonte
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucio Crinò
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Patricia Borges de Souza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Luigi Pasini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabio Nicolini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabrizio Bianchi
- Unit of Cancer Biomarker, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Manel Juan
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Hugo Calderon
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Chiara Magnoni
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luca Gazzola
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paola Ulivi
- Translational Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Massimiliano Mazza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
15
|
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
|
16
|
Huang X, Xu S, An Y, Lv X, Tian L, Xu S, Wang L, Zhu W. Revealing 5-(3,5-difluorobenzyl)-1H-indazole as the active pharmacophore of ALK/ROS1 dual inhibitors through theoretical calculations and biological activity evaluations. Bioorg Chem 2025; 154:108014. [PMID: 39642755 DOI: 10.1016/j.bioorg.2024.108014] [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: 11/12/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
Anaplastic lymphoma kinase (ALK) and tyrosine protein kinase (ROS1) are recognized as driver genes in lung cancer, with dual inhibition of both targets offering a promising approach to enhance therapeutic outcomes in non-small cell lung cancer (NSCLC). Although numerous ALK/ROS1 inhibitors have received FDA approval, detailed research into the essential active structural motifs within these inhibitors remains limited. Addressing this gap, the current study employed computer-aided drug design (CADD) methodologies, incorporating bioisosteric and conformational similarity principles to design and synthesize 31 dual-target 2-morpholinobenzamide derivatives. These derivatives each include the 5-(3,5-difluorobenzyl)-1H-indazole, 4-benzylmorpholine, and thiophene moieties. Based on docking binding energies, we proposed that 5-(3,5-difluorobenzyl)-1H-indazole may represent a key pharmacophore for ALK/ROS1 activity. Subsequent kinase and cellular assays validated this hypothesis, with compound X4 exhibiting optimal inhibitory activity against both ALK and ROS1 kinases and lung cancer cell lines, achieving IC50 values of 0.512 µM (ALK), 0.766 µM (ROS1) and 0.034 ± 0.002 µM (H2228). In vitro antitumor assays demonstrated dose-dependent induction of apoptosis in H2228 cells by X4. Western blot (WB) analysis further confirmed that X4 effectively suppresses the expression of p-ALK and p-ERK. Importantly, X4 exhibited high specificity in targeting ALK and ROS1 across a range of kinases. In an H2228 xenograft model, X4 achieved a tumor inhibition rate of 54.71 %, underscoring its considerable potential in ALK/ROS1 inhibition.
Collapse
Affiliation(s)
- Xiaoling Huang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Shidi Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Yufeng An
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Xinya Lv
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Lulu Tian
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China.
| | - Linxiao Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China.
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China.
| |
Collapse
|
17
|
Bearz A, Bertoli E, Stanzione B, De Carlo E, Del Conte A, Bortolot M, Torresan S, Berto E, Da Ros V, Pelin GM, Fassetta K, Rossetto S, Spina M. EML4-ALK: Update on ALK Inhibitors. Int J Mol Sci 2025; 26:308. [PMID: 39796163 PMCID: PMC11719670 DOI: 10.3390/ijms26010308] [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: 12/04/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Since the discovery of the first-generation ALK inhibitor, many other tyrosine kinase inhibitors have been demonstrated to be effective in the first line or further lines of treatment in patients with advanced non-small cell lung cancer with EMLA4-ALK translocation. This review traces the main milestones in the treatment of ALK-positive metastatic patients and the survival outcomes in the first-line and second-line settings with different ALK inhibitors. It presents the two options available for first-line treatment at the present time: sequencing different ALK inhibitors versus using the most potent inhibitor in front-line treatment. The efficacy outcomes of different ALK inhibitors in the first-line setting; the molecular profile of the disease, including mutation resistances and ALK variants and co-mutations; and patients' co-morbidities and inhibitor toxicities should be taken into account to address the choice of the first-line treatment, as suggested in this review.
Collapse
Affiliation(s)
- Alessandra Bearz
- Centro di Riferimento Oncologico di Aviano (CRO), National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (A.D.C.); (G.M.P.); (S.R.)
| | - Elisa Bertoli
- Centro di Riferimento Oncologico di Aviano (CRO), National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (A.D.C.); (G.M.P.); (S.R.)
| | - Brigida Stanzione
- Centro di Riferimento Oncologico di Aviano (CRO), Department of Medical Oncology, IRCCS, 33081 Aviano, Italy; (B.S.); (E.D.C.); (E.B.); (V.D.R.); (K.F.); (M.S.)
| | - Elisa De Carlo
- Centro di Riferimento Oncologico di Aviano (CRO), Department of Medical Oncology, IRCCS, 33081 Aviano, Italy; (B.S.); (E.D.C.); (E.B.); (V.D.R.); (K.F.); (M.S.)
| | - Alessandro Del Conte
- Centro di Riferimento Oncologico di Aviano (CRO), National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (A.D.C.); (G.M.P.); (S.R.)
| | - Martina Bortolot
- Centro di Riferimento Oncologico di Aviano (CRO), Department of Medical Oncology, IRCCS, 33081 Aviano, Italy; (B.S.); (E.D.C.); (E.B.); (V.D.R.); (K.F.); (M.S.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Sara Torresan
- Centro di Riferimento Oncologico di Aviano (CRO), Department of Medical Oncology, IRCCS, 33081 Aviano, Italy; (B.S.); (E.D.C.); (E.B.); (V.D.R.); (K.F.); (M.S.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Eleonora Berto
- Centro di Riferimento Oncologico di Aviano (CRO), Department of Medical Oncology, IRCCS, 33081 Aviano, Italy; (B.S.); (E.D.C.); (E.B.); (V.D.R.); (K.F.); (M.S.)
| | - Valentina Da Ros
- Centro di Riferimento Oncologico di Aviano (CRO), Department of Medical Oncology, IRCCS, 33081 Aviano, Italy; (B.S.); (E.D.C.); (E.B.); (V.D.R.); (K.F.); (M.S.)
| | - Giulia Maria Pelin
- Centro di Riferimento Oncologico di Aviano (CRO), National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (A.D.C.); (G.M.P.); (S.R.)
| | - Kelly Fassetta
- Centro di Riferimento Oncologico di Aviano (CRO), Department of Medical Oncology, IRCCS, 33081 Aviano, Italy; (B.S.); (E.D.C.); (E.B.); (V.D.R.); (K.F.); (M.S.)
| | - Silvia Rossetto
- Centro di Riferimento Oncologico di Aviano (CRO), National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (A.D.C.); (G.M.P.); (S.R.)
| | - Michele Spina
- Centro di Riferimento Oncologico di Aviano (CRO), Department of Medical Oncology, IRCCS, 33081 Aviano, Italy; (B.S.); (E.D.C.); (E.B.); (V.D.R.); (K.F.); (M.S.)
| |
Collapse
|
18
|
Schneider JL, Han S, Nabel CS. Fuel for thought: targeting metabolism in lung cancer. Transl Lung Cancer Res 2024; 13:3692-3717. [PMID: 39830762 PMCID: PMC11736591 DOI: 10.21037/tlcr-24-662] [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: 07/30/2024] [Accepted: 11/22/2024] [Indexed: 01/22/2025]
Abstract
For over a century, we have appreciated that the biochemical processes through which micro- and macronutrients are anabolized and catabolized-collectively referred to as "cellular metabolism"-are reprogrammed in malignancies. Cancer cells in lung tumors rewire pathways of nutrient acquisition and metabolism to meet the bioenergetic demands for unchecked proliferation. Advances in precision medicine have ushered in routine genotyping of patient lung tumors, enabling a deeper understanding of the contribution of altered metabolism to tumor biology and patient outcomes. This paradigm shift in thoracic oncology has spawned a new enthusiasm for dissecting oncogenotype-specific metabolic phenotypes and creates opportunity for selective targeting of essential tumor metabolic pathways. In this review, we discuss metabolic states across histologic and molecular subtypes of lung cancers and the additional changes in tumor metabolic pathways that occur during acquired therapeutic resistance. We summarize the clinical investigation of metabolism-specific therapies, addressing successes and limitations to guide the evaluation of these novel strategies in the clinic. Beyond changes in tumor metabolism, we also highlight how non-cellular autonomous processes merit particular consideration when manipulating metabolic processes systemically, such as efforts to disentangle how lung tumor cells influence immunometabolism. As the future of metabolic therapeutics hinges on use of models that faithfully recapitulate metabolic rewiring in lung cancer, we also discuss best practices for harmonizing workflows to capture patient specimens for translational metabolic analyses.
Collapse
Affiliation(s)
- Jaime L. Schneider
- Department of Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Christopher S. Nabel
- Department of Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
19
|
Wang KL, Yeh TY, Hsu PC, Wong TH, Liu JR, Chern JW, Lin MH, Yu CW. Discovery of novel anaplastic lymphoma kinase (ALK) and histone deacetylase (HDAC) dual inhibitors exhibiting antiproliferative activity against non-small cell lung cancer. J Enzyme Inhib Med Chem 2024; 39:2318645. [PMID: 38465731 DOI: 10.1080/14756366.2024.2318645] [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: 08/10/2023] [Accepted: 01/11/2024] [Indexed: 03/12/2024] Open
Abstract
A series of novel benzimidazole derivatives were designed and synthesised based on the structures of reported oral available ALK inhibitor and HDAC inhibitor, pracinostat. In enzymatic assays, compound 3b, containing a 2-acyliminobenzimidazole moiety and hydroxamic acid side chain, could inhibit both ALK and HDAC6 (IC50 = 16 nM and 1.03 µM, respectively). Compound 3b also inhibited various ALK mutants known to be involved in crizotinib resistance, including mutant L1196M (IC50, 4.9 nM). Moreover, 3b inhibited the proliferation of several cancer cell lines, including ALK-addicted H2228 cells. To evaluate its potential for treating cancers in vivo, 3b was used in a human A549 xenograft model with BALB/c nude mice. At 20 mg/kg, 3b inhibited tumour growth by 85% yet had a negligible effect on mean body weight. These results suggest a attracting route for the further research and optimisation of dual ALK/HDAC inhibitors.
Collapse
Affiliation(s)
- Kang-Li Wang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsung-Yu Yeh
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Chen Hsu
- Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzu-Hsuan Wong
- Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jia-Rong Liu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ji-Wang Chern
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Miao-Hsia Lin
- Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chao-Wu Yu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
20
|
Cullum S, Vang H, Glover M, Alammarah H, Morton H, Pham N, Rahman M, Khan SA. Case report: Dramatic impact of DNA next generation sequencing results using specific targeted therapies- ALK and PIK3CA. Front Oncol 2024; 14:1462930. [PMID: 39659799 PMCID: PMC11628483 DOI: 10.3389/fonc.2024.1462930] [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: 07/10/2024] [Accepted: 10/21/2024] [Indexed: 12/12/2024] Open
Abstract
In the era of targeted therapies, the clinical importance and utility of next-generation sequencing (NGS) has expanded significantly. Owing to the relative ease and financial feasibility of NGS, the use of personalized treatment strategies has the potential to revolutionize cancer care. In this case report, we explored the use of NGS in salivary gland carcinoma (SGC) and spindle cell neoplasm of the scalp. In our patient with SGC, NGS revealed a GPHN-ALK variant that allowed off-label treatment with alectinib, with a remarkable response in primary and metastatic foci. Similarly, the use of NGS in a cutaneous neoplasm in which no definitive diagnosis could be reached by pathology and which had progressed through standard of care treatment elucidated a PIK3CA mutation in which alpelisib was added and ultimately halted POD. Here, we discuss the use of NGS, future projections, and our recommendations.
Collapse
Affiliation(s)
- Stav Cullum
- Division of Oncology, Department of Medicine, Stanford Cancer Institute and Stanford University, Stanford, CA, United States
| | - Hlu Vang
- Department of Otolaryngology/Head & Neck Surgery Divisions, Stanford University, Stanford, CA, United States
| | - Michael Glover
- Division of Oncology, Department of Medicine, Stanford Cancer Institute and Stanford University, Stanford, CA, United States
| | - Howra Alammarah
- Division of Oncology, Department of Medicine, Stanford Cancer Institute and Stanford University, Stanford, CA, United States
| | - Heather Morton
- Division of Oncology, Department of Medicine, Stanford Cancer Institute and Stanford University, Stanford, CA, United States
| | - Nancy Pham
- Department of Radiology, Stanford University, Stanford, CA, United States
| | - Mobeen Rahman
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Saad A. Khan
- Division of Oncology, Department of Medicine, Stanford Cancer Institute and Stanford University, Stanford, CA, United States
| |
Collapse
|
21
|
Jiang Z, Gu Z, Yu X, Cheng T, Liu B. Research progress on the role of bypass activation mechanisms in resistance to tyrosine kinase inhibitors in non-small cell lung cancer. Front Oncol 2024; 14:1447678. [PMID: 39582541 PMCID: PMC11581962 DOI: 10.3389/fonc.2024.1447678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/25/2024] [Indexed: 11/26/2024] Open
Abstract
The clinical application of small molecule tyrosine kinase inhibitors (TKIs) has significantly improved the quality of life and prognosis of patients with non-small cell lung cancer (NSCLC) carrying driver genes. However, resistance to TKI treatment is inevitable. Bypass signal activation is one of the important reasons for TKI resistance. Although TKI drugs inhibit downstream signaling pathways of driver genes, key signaling pathways within tumor cells can still be persistently activated through bypass routes such as MET gene amplification, EGFR gene amplification, and AXL activation. This continuous activation maintains tumor cell growth and proliferation, leading to TKI resistance. The fundamental strategy to treat TKI resistance mediated by bypass activation involves simultaneously inhibiting the activated bypass signals and the original driver gene signaling pathways. Some clinical trials based on this combined treatment approach have yielded promising preliminary results, offering more treatment options for NSCLC patients with TKI resistance. Additionally, early identification of resistance mechanisms through liquid biopsy, personalized targeted therapy against these mechanisms, and preemptive targeting of drug-tolerant persistent cells may provide NSCLC patients with more sustained and effective treatment.
Collapse
Affiliation(s)
- Ziyang Jiang
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihan Gu
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaomin Yu
- Department of Emergency Medicine, West China Hospital, Sichuan University, West China School of Nursing, Sichuan University, Chengdu, China
- Institute of Disaster Medicine, Sichuan University, Chengdu, China
- Nursing Key Laboratory of Sichuan Province, West China Hospital, Chengdu, China
| | - Tao Cheng
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Bofu Liu
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Chen X, Zhang J, Li B, Yan F. Determining doses for backfill cohorts based on patient-reported outcome. BMC Med Res Methodol 2024; 24:270. [PMID: 39516724 PMCID: PMC11546322 DOI: 10.1186/s12874-024-02398-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Incorporating backfill cohorts in phase I oncology trials is a recently developed strategy for dose optimization. However, the efficacy assessment window is long in general, causing a lag in identifying ineffective doses and more patients being backfilled to those doses. There is necessity to investigate how to use patient-reported outcomes (PRO) to determine doses for backfill cohorts. METHODS We propose a unified Bayesian design framework, called 'Backfill-QoL', to utilize patient-reported quality of life (QoL) data into phase I oncology trials with backfill cohorts, including methods for trial monitoring, algorithm for dose-finding, and criteria for dose selection. Simulation studies and sensitivity analyses are conducted to evaluate the proposed Backfill-QoL design. RESULTS The simulation studies demonstrate that the Backfill-QoL design is more efficient than traditional dose-expansion strategy, and fewer patients would be allocated to doses with unacceptable QoL profiles. A user-friendly Windows desktop application is developed and freely available for implementing the proposed design. CONCLUSIONS The Backfill-QoL design enables continuous monitoring of safety, efficacy and QoL outcomes, and the recommended phase II dose (RP2D) can be identified in a more patient-centered perspective.
Collapse
Affiliation(s)
- Xin Chen
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China
| | - Jingyi Zhang
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China
| | - Bosheng Li
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China
| | - Fangrong Yan
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
23
|
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
|
24
|
Ni H, Wang Z, Tang Y, Lu J, Zhu Z, Qiu Y, Chen Z, Wang Z. Tyrosine kinase inhibitors in the treatment of leptomeningeal carcinomatosis. Cell Biol Int 2024; 48:1450-1462. [PMID: 39136350 DOI: 10.1002/cbin.12230] [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: 12/28/2023] [Revised: 06/28/2024] [Accepted: 07/31/2024] [Indexed: 10/19/2024]
Abstract
Leptomeningeal carcinomatosis (LMC) is a devastating complication of advanced cancers, such as lung cancer and breast cancer, which is usually indicative of a poor prognosis. The current treatments for LMC include palliative care, with others aiming to prolong survival and relieve neurological symptoms. Traditional treatments for LMC include radiotherapy, systemic chemotherapy, and intrathecal injection. Furthermore, the application of molecularly targeted agents, such as antiepidermal growth factor receptor (anti-EGFR), antihuman epidermal growth factor receptor 2 (anti-HER2), and anti-PD-1 monoclonal antibody, have prolonged the survival of LMC patients. Targeted therapy with tyrosine kinase inhibitors has also been proven to be an effective treatment. Tyrosine kinases can be overactive or expressed at high levels in some cancer cells; therefore, the use of tyrosine kinase inhibitors may prevent the activation of tumor-related pathways, preventing cancer cell growth. The EGFR family are cell surface receptors directly related to tumor occurrence with tyrosine kinase activity; it is the most widely used target for tyrosine kinase inhibitors in the treatment of LMC. In this review, we introduced the clinical manifestation and diagnostic criteria of LMC, clarified the treatment mechanism of tyrosine kinase inhibitors for LMC with mutations in EGFR, HER2, or anaplastic lymphoma kinase, reviewed the current application of various generation tyrosine kinase inhibitors in patients with LMC, and discussed new clinical trials and the future directions of tyrosine kinase inhibitor therapy.
Collapse
Affiliation(s)
- Hanyu Ni
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Zilan Wang
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yanbing Tang
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Jiaye Lu
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zixiang Zhu
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Youjia Qiu
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhouqing Chen
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhong Wang
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
25
|
Dong J, Li L, Deng T, Song H, Zhang S, Zhong M. Interstitial lung disease associated with ALK inhibitors and risk factors: an updated comparative pharmacovigilance analysis. Front Pharmacol 2024; 15:1361443. [PMID: 39399468 PMCID: PMC11466793 DOI: 10.3389/fphar.2024.1361443] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Background Inhibitors of the anaplastic lymphoma kinase (ALK) gene mutation are first-line treatments in patients with ALK-positive lung cancer. The FDA label warns of the risk of interstitial lung disease (ILD) in patients receiving ALK TKIs. However, ILD associated with ALK TKIs is not fully understood. The aim of this study was to characterize the features of ALK TKI-related ILD and to explore risk factors for ALK TKI-related ILD. Methods FDA's Adverse Event Reporting System (FAERS) reports from 2011 Q1 to 2023 Q2 were extracted and combined. Standardized MedDRA queries (SMQs) were used to search for AEs at the preferred term (PT) level. Four algorithms were employed to quantify the signals of ILD associated with ALK TKIs. The risk of ILD was further analyzed using logistic regression. Results A total of 20,064 reports of ALK TKIs and 640 (3.2%) reports of ILD AEs were extracted. Significant disproportionality was detected in all five ALK TKIs. Interstitial lung disease and pneumonitis were the most common lung toxicities induced by ALK TKIs. Results of further analyses revealed a different spectrum of lung toxicity among the various TKIs. The median time to onset of ILD related to ALK TKIs was 53 days (Q1:12, Q3:209), and more than 70% of AEs occurred within the first 2 months. Logistic regression analysis and risk prediction model both showed that different ALK TKIs and their combination with PPIs, amlodipine, and magnesium oxide were independent risk factors for ILD (p<0.05). Conclusion ALK TKIs have different safety profiles regarding lung toxicity, which normally occurs within the first 2 months. Administration in combination with PPIs, amlodipine, and magnesium oxide significantly increases the risk of ILD. These results provide risk prediction for ILD related to ALK TKIs and support pharmacovigilance to promote safe prescribing in oncology.
Collapse
Affiliation(s)
- Junli Dong
- Department of Pharmacy, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pharmacy, Wuhan No.1 hospital, Wuhan, China
| | - Lulu Li
- Department of Pharmacy, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pharmacy, Wuhan No.1 hospital, Wuhan, China
| | - Tiying Deng
- Department of Pharmacy, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pharmacy, Wuhan No.1 hospital, Wuhan, China
| | - Haibin Song
- Department of Oncology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Oncology, Wuhan No.1 hospital, Wuhan, China
| | - Shaohui Zhang
- Department of Pharmacy, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pharmacy, Wuhan No.1 hospital, Wuhan, China
| | - Minyu Zhong
- Department of Oncology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Oncology, Wuhan No.1 hospital, Wuhan, China
| |
Collapse
|
26
|
Pfeil AJ, Hale JD, Zhang TS, Wakayama K, Miyazaki I, Odintsov I, Somwar R. Preclinical evaluation of targeted therapies for central nervous system metastases. Dis Model Mech 2024; 17:dmm050836. [PMID: 39344915 PMCID: PMC11463968 DOI: 10.1242/dmm.050836] [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] [Indexed: 10/01/2024] Open
Abstract
The central nervous system (CNS) represents a site of sanctuary for many metastatic tumors when systemic therapies that control the primary tumor cannot effectively penetrate intracranial lesions. Non-small cell lung cancers (NSCLCs) are the most likely of all neoplasms to metastasize to the brain, with up to 60% of patients developing CNS metastases during the disease process. Targeted therapies such as tyrosine kinase inhibitors (TKIs) have helped reduce lung cancer mortality but vary considerably in their capacity to control CNS metastases. The ability of these therapies to effectively target lesions in the CNS depends on several of their pharmacokinetic properties, including blood-brain barrier permeability, affinity for efflux transporters, and binding affinity for both plasma and brain tissue. Despite the existence of numerous preclinical models with which to characterize these properties, many targeted therapies have not been rigorously tested for CNS penetration during the discovery process, whereas some made it through preclinical testing despite poor brain penetration kinetics. Several TKIs have now been engineered with the characteristics of CNS-penetrant drugs, with clinical trials proving these efforts fruitful. This Review outlines the extent and variability of preclinical evidence for the efficacy of NSCLC-targeted therapies, which have been approved by the US Food and Drug Administration (FDA) or are in development, for treating CNS metastases, and how these data correlate with clinical outcomes.
Collapse
Affiliation(s)
- Alexander J. Pfeil
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA
| | - Joshua D. Hale
- University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA
| | - Tiger S. Zhang
- University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA
| | - Kentaro Wakayama
- Taiho Pharmaceutical Co. Ltd. 3, Okubo, Tsukuba, Ibaraki 300-2611, Japan
| | - Isao Miyazaki
- Taiho Pharmaceutical Co. Ltd. 3, Okubo, Tsukuba, Ibaraki 300-2611, Japan
| | - Igor Odintsov
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 021105, USA
| | - Romel Somwar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
27
|
Hu Q, Remsing Rix LL, Desai B, Miroshnychenko D, Li X, Welsh EA, Fang B, Wright GM, Chaudhary N, Kroeger JL, Doebele RC, Koomen JM, Haura EB, Marusyk A, Rix U. Cancer-associated fibroblasts confer ALK inhibitor resistance in EML4-ALK -driven lung cancer via concurrent integrin and MET signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609975. [PMID: 39253447 PMCID: PMC11383036 DOI: 10.1101/2024.08.27.609975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are associated with tumor progression and modulate drug sensitivity of cancer cells. However, the underlying mechanisms are often incompletely understood and crosstalk between tumor cells and CAFs involves soluble secreted as well as adhesion proteins. Interrogating a panel of non-small cell lung cancer (NSCLC) cell lines driven by EML4-ALK fusions, we observed substantial CAF-mediated drug resistance to clinical ALK tyrosine kinase inhibitors (TKIs). Array-based cytokine profiling of fibroblast-derived conditioned- media identified HGF-MET signaling as a major contributor to CAF-mediated paracrine resistance that can be overcome by MET TKIs. However, 'Cell Type specific labeling using Amino acid Precursors' (CTAP)-based expression and phosphoproteomics in direct coculture also highlighted a critical role for the fibronectin-integrin pathway. Flow cytometry analysis confirmed activation of integrin β1 (ITGB1) in lung cancer cells by CAF coculture. Treatment with pharmacological inhibitors, cancer cell-specific silencing or CRISPR-Cas9-mediated knockout of ITGB1 overcame adhesion protein-mediated resistance. Concurrent targeting of MET and integrin signaling effectively abrogated CAF-mediated resistance of EML4-ALK -driven NSCLC cells to ALK TKIs in vitro . Consistently, combination of the ALK TKI alectinib with the MET TKI capmatinib and/or the integrin inhibitor cilengitide was significantly more efficacious than single agent treatment in suppressing tumor growth using an in vivo EML4-ALK -dependent allograft mouse model of NSCLC. In summary, these findings emphasize the complexity of resistance-associated crosstalk between CAFs and cancer cells, which can involve multiple concurrent signaling pathways, and illustrate how comprehensive elucidation of paracrine and juxtacrine resistance mechanisms can inform on more effective therapeutic approaches.
Collapse
|
28
|
Arnaoutakis K, Wan Y, Elliott J, Young M, Yin Y, Leventakos K, Lin HM, Dimou A. Real-World Treatment Patterns and Outcomes Across Three Lines of Therapy in Patients with ALK+ NSCLC. Adv Ther 2024; 41:3217-3231. [PMID: 38916812 PMCID: PMC11263478 DOI: 10.1007/s12325-024-02899-6] [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: 03/14/2024] [Accepted: 05/10/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION Anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKIs) are standard first- and second-line treatment for advanced ALK+ non-small cell lung cancer (NSCLC). We evaluated outcomes in patients with ALK+ NSCLC receiving third-line ALK TKI versus non-ALK-directed therapy. METHODS Flatiron Health OncoEMR data were extracted for patients with ALK+ NSCLC initiating first-line ALK TKI between January 2015 and March 2022 followed by second-line ALK TKI and third-line ALK TKI (group A) or non-TKI therapy (group B). Time-to-treatment discontinuation (TTD) and overall survival (OS) were analyzed using multivariate modelling. RESULTS Among patients receiving third-line ALK TKI (A, n = 85) or non-TKI therapy (B, n = 43), most received first-line crizotinib (A/B: 64%/60%) and second-line alectinib (36%/30%), ceritinib (24%/19%), or lorlatinib (15%/30%). Common third-line treatments were lorlatinib/alectinib (41%/33%) in A and immunotherapy, chemotherapy, or chemotherapy + immunotherapy (30%/28%/21%) in B. Group A versus B had longer TTD of first-line treatment (hazard ratio [HR] 0.62, 95% confidence interval [CI] 0.41-0.93; p = 0.020) and second-line treatment (HR 0.50, 95% CI 0.33-0.75; p < 0.001) and longer OS from start of first-line treatment (HR 0.32, 95% CI 0.19-0.54; p < 0.001) and second-line treatment (HR 0.40, 95% CI 0.24-0.66; p < 0.001). For third-line treatment, median TTD (A/B) was 6.2/2.4 months (HR 0.61, 95% CI 0.37-1.00; p = 0.049) and OS was 17.6/6.5 months (HR 0.57, 95% CI 0.33-0.98; p = 0.042). CONCLUSIONS Patients receiving third-line non-ALK-directed therapy had suboptimal outcomes on prior TKIs. Patients with longer duration of prior ALK TKI treatment appeared to benefit from third-line ALK TKIs.
Collapse
Affiliation(s)
| | - Yin Wan
- Takeda Development Center Americas, Inc., 500 Kendall Street, Cambridge, MA, 02142, USA
| | | | - Matt Young
- Takeda Pharmaceuticals America, Inc., Lexington, MA, USA
| | - Yu Yin
- Takeda Development Center Americas, Inc., 500 Kendall Street, Cambridge, MA, 02142, USA
| | | | - Huamao M Lin
- Global Evidence and Outcomes Research, Takeda Development Center Americas, Inc., 500 Kendall Street, Cambridge, MA, 02142, USA.
| | | |
Collapse
|
29
|
Alotaibi H, Anis AM, Alloghbi A, Alshammari K. Oncology Early-Phase Clinical Trials in the Middle East and North Africa: A Review of the Current Status, Challenges, Opportunities, and Future Directions. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2024; 7:178-189. [PMID: 39219998 PMCID: PMC11361343 DOI: 10.36401/jipo-23-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 09/04/2024]
Abstract
Clinical trials, the empirical discipline of medical experimentation conducted on human subjects, have engendered a paradigm shift in medical research. The need for new clinical studies is paramount in the Middle East and North Africa (MENA) region, with its rising cancer incidence and demand for efficient oncology treatments. This paper comprehensively reviews the challenges, opportunities, and future directions of phase I oncology clinical trials in the MENA region. Early-phase trials are vital in determining drug dosage and assessing toxicity, bridging the gap between preclinical research and clinical practice. Considering the unique landscape of MENA, this review explores regulatory aspects, specific hurdles faced, potential advantages, and areas for improvement in conducting these trials. Various future directions can be pursued to maximize the potential of phase I oncology trials in MENA. While regulatory bodies like the Ministry of Health adhere to the International Conference on Harmonization-Good Clinical Practice guidelines, a unified system meeting high standards would yield better results. Strengthening research infrastructure, establishing research centers, incorporating clinical trial education into the curriculum, and improving access to medical facilities are crucial. Enhancing consumer understanding of research would facilitate increased participation and promote sustainability in trial recruitment. Navigating various funding sources would open the door for more funding opportunities. Collaborations between academia, industry, and regulatory bodies, both international and local, should be fostered to promote knowledge sharing, resource pooling, and harmonization of standards. Such collaborations would contribute to the sustainability of clinical trial activities by leveraging collective expertise, sharing research infrastructure, and distributing the burden of regulatory compliance. By adopting these strategies, the MENA region can advance its capacity to conduct early phases of oncology trials and contribute significantly to the global medical research landscape.
Collapse
Affiliation(s)
- Hawazin Alotaibi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Amna M. Anis
- Biomedical Engineering Department, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
- College of Medicine, Al Faisal University, Riyadh, Saudi Arabia
| | - Abdurahman Alloghbi
- Medical Oncology Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Kanan Alshammari
- Department of Oncology, Ministry of National Guard and Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
30
|
Tobiášová K, Barthová M, Janáková Ľ, Lešková K, Farkašová A, Loderer D, Grendár M, Plank L. Discordant ALK Status in Non-Small Cell Lung Carcinoma: A Detailed Reevaluation Comparing IHC, FISH, and NGS Analyses. Int J Mol Sci 2024; 25:8168. [PMID: 39125737 PMCID: PMC11312000 DOI: 10.3390/ijms25158168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
ALK detection was performed on 2813 EGFR-unmutated NSCLC cases by simultaneous use of immunohistochemistry (VENTANA® anti-ALK D5F3, Roche Molecular Systems, Inc., Rotkreuz, Switzerland) and fluorescence in situ hybridization with the ALK break apart and the ALK/EML4 fusion probe (ZytoVision, Bremerhaven, Germany). A total of 33 cases were positive discordant (FISH-positive, IHC-negative) and 17 cases were negative discordant (FISH-negative, IHC-positive). This study's aim was to reevaluate the methods used and compare discordant samples to positive concordant samples in order to ellucidate the differences. FISH signal variants were examined and compared. Positive discordant cases featured one pattern of ALK rearrangement in 41.4%, two patterns in 48.3%, and three patterns in 10.3% of analysed samples, with a higher variability of detected patterns and a higher number of ALK copy gains. Positive concordant cases displayed one pattern of rearrangement in 82%, two patterns in 17.8%, and three patterns in 0.6% of analysed samples. The association between number of patterns and concordance/discordance was statistically significant (p < 0.05). Eleven positive discordant and two negative concordant cases underwent NGS analysis, which resulted in identification of ALK fusion in one positive discordant and two negative discordant cases. Positive protein expression regardless of FISH result correlated more with a positive NGS result compared to samples with a positive FISH result with negative protein expression. FISH analysis was able to detect atypical or heterogenous patterns of rearrangement in a proportion of cases with negative protein expression, which may be associated with more extensive genetic alterations rather than true ALK rearrangement.
Collapse
Affiliation(s)
- Katarína Tobiášová
- Department of Pathological Anatomy, University Hospital Martin, Jessenius Faculty of Medicine, Comenius University, 036 01 Martin, Slovakia; (K.T.)
| | - Martina Barthová
- Department of Pathological Anatomy, University Hospital Martin, Jessenius Faculty of Medicine, Comenius University, 036 01 Martin, Slovakia; (K.T.)
| | - Ľuboslava Janáková
- Department of Pathological Anatomy, University Hospital Martin, Jessenius Faculty of Medicine, Comenius University, 036 01 Martin, Slovakia; (K.T.)
| | - Katarína Lešková
- Department of Pathological Anatomy, University Hospital Martin, Jessenius Faculty of Medicine, Comenius University, 036 01 Martin, Slovakia; (K.T.)
| | | | - Dušan Loderer
- Biomedical Centre Martin—BioMed Martin, Jessenius Faculty of Medicine, Comenius University, 036 01 Martin, Slovakia
| | - Marián Grendár
- Biomedical Centre Martin—BioMed Martin, Jessenius Faculty of Medicine, Comenius University, 036 01 Martin, Slovakia
| | - Lukáš Plank
- Department of Pathological Anatomy, University Hospital Martin, Jessenius Faculty of Medicine, Comenius University, 036 01 Martin, Slovakia; (K.T.)
- Martin’s Biopsy Center, Ltd., 036 01 Martin, Slovakia
| |
Collapse
|
31
|
Naso JR, Yip S, Hughesman C, Melosky B, Dowhy T, McConechy MK, English JC, Brasher PMA, Choi J, Grant K, Yee J, Lam S, McGuire A. Confirmation of Recurrent Lung Cancer Following Resection Using Liquid Biopsy, a Proof-of-Concept Real-World Study. Curr Oncol 2024; 31:4052-4062. [PMID: 39057174 PMCID: PMC11276127 DOI: 10.3390/curroncol31070302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Appropriate management requires timely and accurate confirmation of non-small cell lung cancer (NSCLC) recurrence in patients who have had curative-intent surgical resection. We assessed the association between circulating tumor DNA (ctDNA) identified using amplicon sequencing and evidence of recurrence on CT surveillance. A prospective cohort study of NSCLC patients with early-stage disease undergoing curative-intent resection was conducted. Surveillance was performed post-operatively at pre-defined intervals with both liquid biopsy and chest CT imaging. Amplicon panel next-generation sequencing was performed on DNA and RNA from tumor tissue and on plasma cell-free DNA for tumor-informed ctDNA detection. Resected tumors from 78 NSCLC patients were analyzed. Alterations were detected on the DNA assay for 65 tumors and only on the RNA assay for 4 tumors. Of the 65 patients with alterations detected on the tumor DNA assay, 29 completed post-operative liquid biopsy testing. Four of those 29 patients had evidence of recurrence on imaging, of whom two had biopsy confirmation of recurrence and detectable ctDNA at the 12-month follow-up. Molecular confirmation of NSCLC recurrence can be provided through amplicon sequencing of plasma cell-free DNA in cases with imaging evidence of recurrence. Invasive tissue diagnosis may be avoidable in patients with ctDNA confirmation of recurrence that is suspected based on imaging. Further study of ctDNA assessment technologies in the setting of suspected recurrence is necessary to inform post-operative lung cancer surveillance guidelines.
Collapse
Affiliation(s)
- Julia R. Naso
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V5Z 4E6, Canada; (J.R.N.)
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V5Z 4E6, Canada; (J.R.N.)
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
- Cancer Genetics and Genomics Laboratory, BC Cancer, Vancouver, BC V5Z 4E6, Canada
- Department of Pathology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - Curtis Hughesman
- Department of Pathology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - Barb Melosky
- Department of Medical Oncology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - Tanner Dowhy
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, BC V5Z 4S6, Canada
| | | | - John C. English
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
| | - Penelope M. A. Brasher
- Centre for Clinical Epidemiology and Evaluation, Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| | - James Choi
- Division of Thoracic Surgery, Department of Surgery, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
| | - Kyle Grant
- Division of Thoracic Surgery, Department of Surgery, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
| | - John Yee
- Division of Thoracic Surgery, Department of Surgery, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
| | - Stephen Lam
- Division of Respirology, Department of Medicine, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - Anna McGuire
- Division of Thoracic Surgery, Department of Surgery, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
- Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
32
|
Boileve A, Smolenschi C, Lambert A, Boige V, Tarabay A, Valery M, Fuerea A, Pudlarz T, Conroy T, Hollebecque A, Ducreux M. Role of molecular biology in the management of pancreatic cancer. World J Gastrointest Oncol 2024; 16:2902-2914. [PMID: 39072173 PMCID: PMC11271790 DOI: 10.4251/wjgo.v16.i7.2902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/04/2024] [Accepted: 05/21/2024] [Indexed: 07/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents significant challenges in patient management due to a dismal prognosis, increasing incidence, and limited treatment options. In this regard, precision medicine, which personalizes treatments based on tumour molecular characteristics, has gained great interest. However, its widespread implementation is not fully endorsed in current recommendations. This review explores key molecular alterations in PDAC, while emphasizing differences between KRAS-mutated and KRAS-wild-type tumours. It assesses the practical application of precision medicine in clinical settings and outlines potential future directions with respect to PDAC. Actionable molecular targets are examined with the aim of enhancing our understanding of PDAC molecular biology. Insights from this analysis may contribute to a more refined and personalized approach to pancreatic cancer treatment, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Alice Boileve
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | | | - Aurélien Lambert
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Nancy 54519, France
| | - Valérie Boige
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Anthony Tarabay
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Marine Valery
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Alina Fuerea
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Thomas Pudlarz
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Thierry Conroy
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Nancy 54519, France
| | | | - Michel Ducreux
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| |
Collapse
|
33
|
Pliszka M, Szablewski L. Associations between Diabetes Mellitus and Selected Cancers. Int J Mol Sci 2024; 25:7476. [PMID: 39000583 PMCID: PMC11242587 DOI: 10.3390/ijms25137476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Cancer is one of the major causes of mortality and is the second leading cause of death. Diabetes mellitus is a serious and growing problem worldwide, and its prevalence continues to grow; it is the 12th leading cause of death. An association between diabetes mellitus and cancer has been suggested for more than 100 years. Diabetes is a common disease diagnosed among patients with cancer, and evidence indicates that approximately 8-18% of patients with cancer have diabetes, with investigations suggesting an association between diabetes and some particular cancers, increasing the risk for developing cancers such as pancreatic, liver, colon, breast, stomach, and a few others. Breast and colorectal cancers have increased from 20% to 30% and there is a 97% increased risk of intrahepatic cholangiocarcinoma or endometrial cancer. On the other hand, a number of cancers and cancer therapies increase the risk of diabetes mellitus. Complications due to diabetes in patients with cancer may influence the choice of cancer therapy. Unfortunately, the mechanisms of the associations between diabetes mellitus and cancer are still unknown. The aim of this review is to summarize the association of diabetes mellitus with selected cancers and update the evidence on the underlying mechanisms of this association.
Collapse
Affiliation(s)
- Monika Pliszka
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| | - Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| |
Collapse
|
34
|
Kim M. Mitochondria of T Lymphocytes Promote Anti-Pulmonary Tumor Immune Response. World J Oncol 2024; 15:472-481. [PMID: 38751696 PMCID: PMC11092414 DOI: 10.14740/wjon1841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/30/2024] [Indexed: 05/18/2024] Open
Abstract
Background B-cell lymphoma 2 (Bcl-2), a protein involved in apoptosis, has been proven to have carcinogenic potential and is well documented. With the recent advancement in optical technology, it has become possible to observe subcellular organelles such as mitochondria in real-time without the need for staining. Consequently, we have examined the movement of mitochondria in cancer cells, correlating it with the regulation of Bcl-2. Methods Using a tomographic microscope, which can detect the internal structure of cells, we observed lung tumor cells. Cells were exposed to a laser beam (λ = 520 nm) inclined at 45°, and holographic images were recorded up to a depth of 30 µm of reconstruction. Results Intriguingly, lung tumor cells rapidly expelled mitochondria upon the attachment of Bcl-2 or B-cell lymphoma extra-large (Bcl-xL) inhibitors. On the other hand, we observed that tumor cells hijack mitochondria from T cells. The hijacked mitochondria were not immediately linked to tumor cell death, but they played a role in assisting granzyme B-induced tumor cell death. Due to lower levels of Bcl-2 and Bcl-xL on the mitochondria of T cells compared to lung tumor cells, immune cells depleted of Bcl-2 and Bcl-xL were co-cultured with the tumor cells. Conclusions As a result, a more effective tumor cell death induced by granzyme B was observed. Additionally, further enhanced anticancer immune response was observed in vivo. Together, we show that modified mitochondria of T cells can provide potential novel strategies towards tumor cell death.
Collapse
Affiliation(s)
- Minsuk Kim
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul 07804, Korea.
| |
Collapse
|
35
|
Tran TO, Vo TH, Le NQK. Omics-based deep learning approaches for lung cancer decision-making and therapeutics development. Brief Funct Genomics 2024; 23:181-192. [PMID: 37519050 DOI: 10.1093/bfgp/elad031] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/04/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023] Open
Abstract
Lung cancer has been the most common and the leading cause of cancer deaths globally. Besides clinicopathological observations and traditional molecular tests, the advent of robust and scalable techniques for nucleic acid analysis has revolutionized biological research and medicinal practice in lung cancer treatment. In response to the demands for minimally invasive procedures and technology development over the past decade, many types of multi-omics data at various genome levels have been generated. As omics data grow, artificial intelligence models, particularly deep learning, are prominent in developing more rapid and effective methods to potentially improve lung cancer patient diagnosis, prognosis and treatment strategy. This decade has seen genome-based deep learning models thriving in various lung cancer tasks, including cancer prediction, subtype classification, prognosis estimation, cancer molecular signatures identification, treatment response prediction and biomarker development. In this study, we summarized available data sources for deep-learning-based lung cancer mining and provided an update on recent deep learning models in lung cancer genomics. Subsequently, we reviewed the current issues and discussed future research directions of deep-learning-based lung cancer genomics research.
Collapse
Affiliation(s)
- Thi-Oanh Tran
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, No 250 Wuxing Street, 110, Taipei, Taiwan
- AIBioMed Research Group, Taipei Medical University, No 250 Wuxing Street, 110, Taipei, Taiwan
- Hematology and Blood Transfusion Center, Bach Mai Hospital, No 78 Giai Phong Street, Hanoi, Viet Nam
| | - Thanh Hoa Vo
- Department of Science, School of Science and Computing, South East Technological University, Waterford X91 K0EK, Ireland
- Pharmaceutical and Molecular Biotechnology Research Center (PMBRC), South East Technological University, Waterford X91 K0EK, Ireland
| | - Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, 110, Taipei, Taiwan
- AIBioMed Research Group, Taipei Medical University, No 250 Wuxing Street, 110, Taipei, Taiwan
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, 250 Wuxing Street, 110, Taipei, Taiwan
- Translational Imaging Research Center, Taipei Medical University Hospital, 252 Wuxing Street, 110, Taipei, Taiwan
| |
Collapse
|
36
|
Hussain S, Mursal M, Verma G, Hasan SM, Khan MF. Targeting oncogenic kinases: Insights on FDA approved tyrosine kinase inhibitors. Eur J Pharmacol 2024; 970:176484. [PMID: 38467235 DOI: 10.1016/j.ejphar.2024.176484] [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: 09/24/2023] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
Protein kinases play pivotal roles in various biological functions, influencing cell differentiation, promoting survival, and regulating the cell cycle. The disruption of protein kinase activity is intricately linked to pathways in tumor development. This manuscript explores the transformative impact of protein kinase inhibitors on cancer therapy, particularly their efficacy in cases driven by targeted mutations. Focusing on key tyrosine kinase inhibitors (TKIs) like Bcr-Abl, Epidermal Growth Factor Receptor (EGFR), and Vascular Endothelial Growth Factor Receptor (VEGFR), it targets critical kinase families in cancer progression. Clinical trial details of these TKIs offer insights into their therapeutic potentials. Learning from FDA-approved kinase inhibitors, the review dissects trends in kinase drug development since imatinib's paradigm-shifting approval in 2001. TKIs have evolved into pivotal drugs, extending beyond oncology. Ongoing clinical trials explore novel kinase targets, revealing the vast potential within the human kinome. The manuscript provides a detailed analysis of advancements until 2022, discussing the roles of specific oncogenic protein kinases in cancer development and carcinogenesis. Our exploration on PubMed for relevant and significant TKIs undergoing pre-FDA approval phase III clinical trials enriches the discussion with valuable findings. While kinase inhibitors exhibit lower toxicity than traditional chemotherapy in cancer treatment, challenges like resistance and side effects emphasize the necessity of understanding resistance mechanisms, prompting the development of novel inhibitors like osimertinib targeting specific mutant proteins. The review advocates thorough research on effective combination therapies, highlighting the future development of more selective RTKIs to optimize patient-specific cancer treatment and reduce adverse events.
Collapse
Affiliation(s)
- Sahil Hussain
- Faculty of Pharmacy, Integral University, Kursi Road, Lucknow, 226026, India
| | - Mohd Mursal
- Faculty of Pharmacy, Integral University, Kursi Road, Lucknow, 226026, India
| | - Garima Verma
- RWE Specialist, HealthPlix Technologies, Bengaluru, Karnataka 560103, India
| | - Syed Misbahul Hasan
- Faculty of Pharmacy, Integral University, Kursi Road, Lucknow, 226026, India
| | - Mohemmed Faraz Khan
- Faculty of Pharmacy, Integral University, Kursi Road, Lucknow, 226026, India.
| |
Collapse
|
37
|
Lara MS, Blakely CM, Riess JW. Targeting MEK in non-small cell lung cancer. Curr Probl Cancer 2024; 49:101065. [PMID: 38341356 DOI: 10.1016/j.currproblcancer.2024.101065] [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: 01/06/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024]
Abstract
The mitogen-activated protein kinase (MAPK or MEK) pathway modulates tumor cell survival and proliferation in non-small cell lung cancer (NSCLC). Unlike RAS or EGFR, activating mutations in MEK are exceedingly rare in NSCLC. Instead, enhanced activation of the MEK pathway is often linked to increased signaling by upstream oncogenic driver mutations. Thus far, MEK inhibitor monotherapy has shown little promise. However, treatment strategies involving MEK inhibition in combination with other targeted therapies in other oncogene-driven NSCLC has proven to be encouraging. For example, MEK inhibition - when combined with BRAF inhibition, - has shown strong anti-tumor activity in BRAF V600 mutated NSCLC. In this review, recent data on MEK inhibitor strategies in NSCLC are summarized. Furthermore, ongoing early phase trials investigating MEK inhibitor combination therapy with immunotherapy, chemotherapy and other oncogene drivers are highlighted. These and other studies could help inform future rational combination strategies of MEK-ERK inhibition in oncogene-driven NSCLC.
Collapse
Affiliation(s)
- Matthew S Lara
- University of California Davis Comprehensive Cancer Center and the UC Davis School of Medicine, Sacramento CA, USA
| | - Collin M Blakely
- University of California San Francisco Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Jonathan W Riess
- University of California Davis Comprehensive Cancer Center and the UC Davis School of Medicine, Sacramento CA, USA.
| |
Collapse
|
38
|
Zheng J, Wang T, Yang Y, Huang J, Feng J, Zhuang W, Chen J, Zhao J, Zhong W, Zhao Y, Zhang Y, Song Y, Hu Y, Yu Z, Gong Y, Chen Y, Ye F, Zhang S, Cao L, Fan Y, Wu G, Guo Y, Zhou C, Ma K, Fang J, Feng W, Liu Y, Zheng Z, Li G, Wang H, Cang S, Wu N, Song W, Liu X, Zhao S, Ding L, Selvaggi G, Wang Y, Xiao S, Wang Q, Shen Z, Zhou J, Zhou J, Zhang L. Updated overall survival and circulating tumor DNA analysis of ensartinib for crizotinib-refractory ALK-positive NSCLC from a phase II study. Cancer Commun (Lond) 2024; 44:455-468. [PMID: 38421881 PMCID: PMC11024683 DOI: 10.1002/cac2.12524] [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: 07/20/2023] [Revised: 12/13/2023] [Accepted: 02/03/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND The initial phase II stuty (NCT03215693) demonstrated that ensartinib has shown clinical activity in patients with advanced crizotinib-refractory, anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer (NSCLC). Herein, we reported the updated data on overall survival (OS) and molecular profiling from the initial phase II study. METHODS In this study, 180 patients received 225 mg of ensartinib orally once daily until disease progression, death or withdrawal. OS was estimated by Kaplan‒Meier methods with two-sided 95% confidence intervals (CIs). Next-generation sequencing was employed to explore prognostic biomarkers based on plasma samples collected at baseline and after initiating ensartinib. Circulating tumor DNA (ctDNA) was detected to dynamically monitor the genomic alternations during treatment and indicate the existence of molecular residual disease, facilitating improvement of clinical management. RESULTS At the data cut-off date (August 31, 2022), with a median follow-up time of 53.2 months, 97 of 180 (53.9%) patients had died. The median OS was 42.8 months (95% CI: 29.3-53.2 months). A total of 333 plasma samples from 168 patients were included for ctDNA analysis. An inferior OS correlated significantly with baseline ALK or tumor protein 53 (TP53) mutation. In addition, patients with concurrent TP53 mutations had shorter OS than those without concurrent TP53 mutations. High ctDNA levels evaluated by variant allele frequency (VAF) and haploid genome equivalents per milliliter of plasma (hGE/mL) at baseline were associated with poor OS. Additionally, patients with ctDNA clearance at 6 weeks and slow ascent growth had dramatically longer OS than those with ctDNA residual and fast ascent growth, respectively. Furthermore, patients who had a lower tumor burden, as evaluated by the diameter of target lesions, had a longer OS. Multivariate Cox regression analysis further uncovered the independent prognostic values of bone metastases, higher hGE, and elevated ALK mutation abundance at 6 weeks. CONCLUSION Ensartinib led to a favorable OS in patients with advanced, crizotinib-resistant, and ALK-positive NSCLC. Quantification of ctDNA levels also provided valuable prognostic information for risk stratification.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Respiratory DiseaseThoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Provincial Clinical Research Center for Respiratory DiseaseHangzhouZhejiangP. R. China
| | - Tao Wang
- Hangzhou Repugene Technology Co., LtdHangzhouZhejiangP. R. China
| | - Yunpeng Yang
- Department of Medical OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongP. R. China
| | - Jie Huang
- Department of Medical OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongP. R. China
| | - Jifeng Feng
- Department of Medical OncologyJiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| | - Wu Zhuang
- Department of Thoracic OncologyFujian Provincial Cancer HospitalFujian Medical University Cancer HospitalFuzhouFujianP. R. China
| | - Jianhua Chen
- Department of Medical Oncology‐ChestHunan Cancer HospitalChangshaHunanP. R. China
| | - Jun Zhao
- Department of Thoracic OncologyBeijing Cancer HospitalBeijingP. R. China
| | - Wei Zhong
- Department of Pulmonary MedicinePeking Union Medical College HospitalChinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingP. R. China
| | - Yanqiu Zhao
- Respiratory Department of Internal MedicineHenan Provincial Cancer HospitalAffiliated Cancer Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Yiping Zhang
- Thoracic Medical OncologyZhejiang Cancer HospitalHangzhouZhejiangP. R. China
| | - Yong Song
- Division of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingJiangsuP. R. China
| | - Yi Hu
- Department of OncologyChinese People's Liberation Army (PLA) General HospitalBeijingP. R. China
| | - Zhuang Yu
- Department of OncologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongP. R. China
| | - Youling Gong
- Department of Thoracic OncologyCancer Center, West China HospitalSichuan UniversityChengduSichuanP. R. China
| | - Yuan Chen
- Department of OncologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Feng Ye
- Department of Medical OncologyCancer HospitalThe First Affiliated Hospital of Xiamen UniversitySchool of Medicine, Xiamen University, Teaching Hospital of Fujian Medical UniversityXiamenFujianP. R. China
| | - Shucai Zhang
- Department of Medical OncologyBeijing Chest HospitalCapital Medical University, Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingP. R. China
| | - Lejie Cao
- Respiratory MedicineThe First Affiliated Hospital of the University of Science and Technology of ChinaAnhui Provincial HospitalHefeiAnhuiP. R. China
| | - Yun Fan
- Thoracic Medical OncologyZhejiang Cancer HospitalHangzhouZhejiangP. R. China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Yubiao Guo
- Pulmonary & Critical Care Medicine, The First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
| | - Chengzhi Zhou
- Respiratory Medicine DepartmentState Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongP. R. China
| | - Kewei Ma
- Cancer Center, The First Hospital of Jilin UniversityChangchunJilinP. R. China
| | - Jian Fang
- Department of Thoracic OncologyBeijing Cancer HospitalBeijingP. R. China
| | - Weineng Feng
- Department of Head and Neck and Thoracic Medical OncologyThe First People's Hospital of FoshanFoshanGuangdongP. R. China
| | - Yunpeng Liu
- Oncology MedicineThe First Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Zhendong Zheng
- Oncology DepartmentGeneral Hospital of Northern Theater CommandShenyangLiaoningP. R. China
| | - Gaofeng Li
- 2nd Department of Thoracic SurgeryYunnan Cancer HospitalKunmingYunnanP. R. China
| | - Huijie Wang
- Medical OncologyFudan University Shanghai Cancer CenterShanghaiShanghaiP. R. China
| | - Shundong Cang
- Medical OncologyHenan Province Peoples HospitalZhengzhouHenanP. R. China
| | - Ning Wu
- PET‐CT Center & Department of Diagnostic RadiologyNational Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingP. R. China
| | - Wei Song
- Department of RadiologyPeking Union Medical College HospitalChinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingP. R. China
| | - Xiaoqing Liu
- Department of Pulmonary OncologyThe Fifth Medical Centre Chinese PLA General HospitalBeijingP. R. China
| | - Shijun Zhao
- Department of Diagnostic RadiologyNational Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingP. R. China
| | - Lieming Ding
- Betta Pharmaceuticals Co., LtdHangzhouZhejiangP. R. China
| | | | - Yang Wang
- Betta Pharmaceuticals Co., LtdHangzhouZhejiangP. R. China
| | - Shanshan Xiao
- Hangzhou Repugene Technology Co., LtdHangzhouZhejiangP. R. China
| | - Qian Wang
- Hangzhou Repugene Technology Co., LtdHangzhouZhejiangP. R. China
| | - Zhilin Shen
- Betta Pharmaceuticals Co., LtdHangzhouZhejiangP. R. China
| | - Jianya Zhou
- Department of Respiratory DiseaseThoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Provincial Clinical Research Center for Respiratory DiseaseHangzhouZhejiangP. R. China
| | - Jianying Zhou
- Department of Respiratory DiseaseThoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Provincial Clinical Research Center for Respiratory DiseaseHangzhouZhejiangP. R. China
| | - Li Zhang
- Department of Medical OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongP. R. China
| |
Collapse
|
39
|
Li S, Zhang H, Chen T, Zhang X, Shang G. Current treatment and novel insights regarding ROS1-targeted therapy in malignant tumors. Cancer Med 2024; 13:e7201. [PMID: 38629293 PMCID: PMC11022151 DOI: 10.1002/cam4.7201] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 03/22/2024] [Accepted: 04/06/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND The proto-oncogene ROS1 encodes an intrinsic type I membrane protein of the tyrosine kinase/insulin receptor family. ROS1 facilitates the progression of various malignancies via self-mutations or rearrangements. Studies on ROS1-directed tyrosine kinase inhibitors have been conducted, and some have been approved by the FDA for clinical use. However, the adverse effects and mechanisms of resistance associated with ROS1 inhibitors remain unknown. In addition, next-generation ROS1 inhibitors, which have the advantage of treating central nervous system metastases and alleviating endogenous drug resistance, are still in the clinical trial stage. METHOD In this study, we searched relevant articles reporting the mechanism and clinical application of ROS1 in recent years; systematically reviewed the biological mechanisms, diagnostic methods, and research progress on ROS1 inhibitors; and provided perspectives for the future of ROS1-targeted therapy. RESULTS ROS1 is most expressed in malignant tumours. Only a few ROS1 kinase inhibitors are currently approved for use in NSCLC, the efficacy of other TKIs for NSCLC and other malignancies has not been ascertained. There is no effective standard treatment for adverse events or resistance to ROS1-targeted therapy. Next-generation TKIs appear capable of overcoming resistance and delaying central nervous system metastasis, but with a greater incidence of adverse effects. CONCLUSIONS Further research on next-generation TKIs regarding the localization of ROS1 and its fusion partners, binding sites for targeted drugs, and coadministration with other drugs is required. The correlation between TKIs and chemotherapy or immunotherapy in clinical practice requires further study.
Collapse
Affiliation(s)
- Shizhe Li
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - He Zhang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Ting Chen
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Xiaowen Zhang
- Medical Research CenterShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Guanning Shang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| |
Collapse
|
40
|
Liu W, Huo G, Chen P. Cost-effectiveness of first-line versus second-line use of brigatinib followed by lorlatinib in patients with ALK-positive non-small cell lung cancer. Front Public Health 2024; 12:1213318. [PMID: 38435286 PMCID: PMC10906082 DOI: 10.3389/fpubh.2024.1213318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Background The ALTA-1 L trial and EXP-3B arm of NCT01970865 trial found that both brigatinib and lorlatinib showed durable and robust responses in treating ALK-positive non-small cell lung cancer (NSCLC) patients. However, brigatinib and lorlatinib treatments are costly and need indefinite administration until the disease progression. Thus, it remains uncertain whether using brigatinib followed by lorlatinib before chemotherapy is cost-effective compared to reserving these two drugs until progression after chemotherapy. Methods We used a Markov model to assess clinical outcomes and healthcare costs of treating ALK-positive NSCLC individuals with brigatinib followed by lorlatinib before chemotherapy versus a strategy of reserving these drugs until progression after chemotherapy. Transition probabilities were estimated using parametric survival modeling based on multiple clinical trials. The drug acquisition costs, adverse events costs, administration costs were extracted from published studies before and publicly available data. We calculated lifetime direct healthcare costs, quality-adjusted life-years (QALYs), and incremental cost-effectiveness ratios from the perspective of a United States payer. Results Our base-case analysis indicated that the incremental cost-effectiveness ratios of using first-line brigatinib followed by lorlatinib compared with second-line brigatinib followed by lorlatinib is $-400,722.09/QALY which meant that second-line brigatinib followed by lorlatinib had less costs and better outcomes. Univariate sensitivity analysis indicated the results were most sensitive to the cost of brigatinib. Probability sensitivity analysis revealed that using brigatinib followed by lorlatinib before chemotherapy had a 0% probability of cost-effectiveness versus delaying these two drugs until progression after chemotherapy at a willingness-to-pay threshold of $150,000 per QALY. Sensitivity analyses conducted revealed the robustness of this result, as incremental cost-effectiveness ratios never exceeded the willingness-to-pay threshold. Conclusion Using brigatinib as first-line treatment followed by lorlatinib for ALK-positive NSCLC may not be cost-effective given current pricing from the perspective of a United States payer. Delaying brigatinib followed by lorlatinib until subsequent lines of treatment may be a reasonable strategy that could limit healthcare costs without affecting clinical outcomes. More mature data are needed to better estimate cost-effectiveness in this setting.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Gengwei Huo
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Peng Chen
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
41
|
Myint KZ, Balasubramanian B, Venkatraman S, Phimsen S, Sripramote S, Jantra J, Choeiphuk C, Mingphruedhi S, Muangkaew P, Rungsakulkij N, Tangtawee P, Suragul W, Farquharson WV, Wongprasert K, Chutipongtanate S, Sanvarinda P, Ponpuak M, Poungvarin N, Janvilisri T, Suthiphongchai T, Yacqub-Usman K, Grabowska AM, Bates DO, Tohtong R. Therapeutic Implications of Ceritinib in Cholangiocarcinoma beyond ALK Expression and Mutation. Pharmaceuticals (Basel) 2024; 17:197. [PMID: 38399413 PMCID: PMC10892566 DOI: 10.3390/ph17020197] [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: 01/08/2024] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a difficult-to-treat cancer, with limited therapeutic options and surgery being the only curative treatment. Standard chemotherapy involves gemcitabine-based therapies combined with cisplatin, oxaliplatin, capecitabine, or 5-FU with a dismal prognosis for most patients. Receptor tyrosine kinases (RTKs) are aberrantly expressed in CCAs encompassing potential therapeutic opportunity. Hence, 112 RTK inhibitors were screened in KKU-M213 cells, and ceritinib, an approved targeted therapy for ALK-fusion gene driven cancers, was the most potent candidate. Ceritinib's cytotoxicity in CCA was assessed using MTT and clonogenic assays, along with immunofluorescence, western blot, and qRT-PCR techniques to analyze gene expression and signaling changes. Furthermore, the drug interaction relationship between ceritinib and cisplatin was determined using a ZIP synergy score. Additionally, spheroid and xenograft models were employed to investigate the efficacy of ceritinib in vivo. Our study revealed that ceritinib effectively killed CCA cells at clinically relevant plasma concentrations, irrespective of ALK expression or mutation status. Ceritinib modulated multiple signaling pathways leading to the inhibition of the PI3K/Akt/mTOR pathway and activated both apoptosis and autophagy. Additionally, ceritinib and cisplatin synergistically reduced CCA cell viability. Our data show ceritinib as an effective treatment of CCA, which could be potentially explored in the other cancer types without ALK mutations.
Collapse
Affiliation(s)
- Kyaw Zwar Myint
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (K.Z.M.); (B.B.); (S.V.); (T.J.)
| | - Brinda Balasubramanian
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (K.Z.M.); (B.B.); (S.V.); (T.J.)
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Simran Venkatraman
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (K.Z.M.); (B.B.); (S.V.); (T.J.)
| | - Suchada Phimsen
- Department of Biochemistry, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (S.P.); (C.C.)
| | - Supisara Sripramote
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.S.); (J.J.); (T.S.)
| | - Jeranan Jantra
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.S.); (J.J.); (T.S.)
| | - Chaiwat Choeiphuk
- Department of Biochemistry, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (S.P.); (C.C.)
| | - Somkit Mingphruedhi
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Paramin Muangkaew
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Narongsak Rungsakulkij
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Pongsatorn Tangtawee
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Wikran Suragul
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Watoo Vassanasiri Farquharson
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Kanokpan Wongprasert
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Somchai Chutipongtanate
- Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Pimtip Sanvarinda
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Marisa Ponpuak
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Naravat Poungvarin
- Department of Clinical Pathology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Tavan Janvilisri
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (K.Z.M.); (B.B.); (S.V.); (T.J.)
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.S.); (J.J.); (T.S.)
| | - Tuangporn Suthiphongchai
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.S.); (J.J.); (T.S.)
| | - Kiren Yacqub-Usman
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (K.Y.-U.); (A.M.G.); (D.O.B.)
| | - Anna M. Grabowska
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (K.Y.-U.); (A.M.G.); (D.O.B.)
| | - David O. Bates
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (K.Y.-U.); (A.M.G.); (D.O.B.)
| | - Rutaiwan Tohtong
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.S.); (J.J.); (T.S.)
| |
Collapse
|
42
|
Chen N, Tyler LC, Le AT, Welsh EA, Fang B, Elliott A, Davies KD, Danhorn T, Riely GJ, Ladanyi M, Haura EB, Doebele RC. MIG6 Mediates Adaptive and Acquired Resistance to ALK/ROS1 Fusion Kinase Inhibition through EGFR Bypass Signaling. Mol Cancer Ther 2024; 23:92-105. [PMID: 37748191 PMCID: PMC10762338 DOI: 10.1158/1535-7163.mct-23-0218] [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: 04/11/2023] [Revised: 08/10/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023]
Abstract
Despite the initial benefit from tyrosine kinase inhibitors (TKI) targeting oncogenic ALK and ROS1 gene fusions in non-small cell lung cancer, complete responses are rare and resistance ultimately emerges from residual tumor cells. Although several acquired resistance mechanisms have been reported at the time of disease progression, adaptative resistance mechanisms that contribute to residual diseases before the outgrowth of tumor cells with acquired resistance are less clear. For the patients who have progressed after TKI treatments, but do not demonstrate ALK/ROS1 kinase mutations, there is a lack of biomarkers to guide effective treatments. Herein, we found that phosphorylation of MIG6, encoded by the ERRFI1 gene, was downregulated by ALK/ROS1 inhibitors as were mRNA levels, thus potentiating EGFR activity to support cell survival as an adaptive resistance mechanism. MIG6 downregulation was sustained following chronic exposure to ALK/ROS1 inhibitors to support the establishment of acquired resistance. A higher ratio of EGFR to MIG6 expression was found in ALK TKI-treated and ALK TKI-resistant tumors and correlated with the poor responsiveness to ALK/ROS1 inhibition in patient-derived cell lines. Furthermore, we identified and validated a MIG6 EGFR-binding domain truncation mutation in mediating resistance to ROS1 inhibitors but sensitivity to EGFR inhibitors. A MIG6 deletion was also found in a patient after progressing to ROS1 inhibition. Collectively, this study identifies MIG6 as a novel regulator for EGFR-mediated adaptive and acquired resistance to ALK/ROS1 inhibitors and suggests EGFR to MIG6 ratios and MIG6-damaging alterations as biomarkers to predict responsiveness to ALK/ROS1 and EGFR inhibitors.
Collapse
Affiliation(s)
- Nan Chen
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Logan C. Tyler
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anh T. Le
- Cell Technologies Shared Resources, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Eric A. Welsh
- Biostatistics and Bioinformatics Shared Resources, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Bin Fang
- Proteomics & Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Andrew Elliott
- Clinical and Translational Research, Caris Life Sciences, Phoenix, Arizona
| | - Kurtis D. Davies
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Thomas Danhorn
- Department of Pharmacology and of University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Gregory J. Riely
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric B. Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Robert C. Doebele
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
43
|
Srivastava S, Jayaswal N, Kumar S, Sharma PK, Behl T, Khalid A, Mohan S, Najmi A, Zoghebi K, Alhazmi HA. Unveiling the potential of proteomic and genetic signatures for precision therapeutics in lung cancer management. Cell Signal 2024; 113:110932. [PMID: 37866667 DOI: 10.1016/j.cellsig.2023.110932] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/15/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
Lung cancer's enduring global significance necessitates ongoing advancements in diagnostics and therapeutics. Recent spotlight on proteomic and genetic biomarker research offers a promising avenue for understanding lung cancer biology and guiding treatments. This review elucidates genetic and proteomic lung cancer biomarker progress and their treatment implications. Technological strides in mass spectrometry-based proteomics and next-generation sequencing enable pinpointing of genetic abnormalities and abnormal protein expressions, furnishing vital data for precise diagnosis, patient classification, and customized treatments. Biomarker-driven personalized medicine yields substantial treatment improvements, elevating survival rates and minimizing adverse effects. Integrating omics data (genomics, proteomics, etc.) enhances understanding of lung cancer's intricate biological milieu, identifying novel treatment targets and biomarkers, fostering precision medicine. Liquid biopsies, non-invasive tools for real-time treatment monitoring and early resistance detection, gain popularity, promising enhanced management and personalized therapy. Despite advancements, biomarker repeatability and validation challenges persist, necessitating interdisciplinary efforts and large-scale clinical trials. Integrating artificial intelligence and machine learning aids analyzing vast omics datasets and predicting treatment responses. Single-cell omics reveal cellular connections and intratumoral heterogeneity, valuable for combination treatments. Biomarkers enable accurate diagnosis, tailored medicines, and treatment response tracking, significantly impacting personalized lung cancer care. This approach spurs patient-centered trials, empowering active patient engagement. Lung cancer proteomic and genetic biomarkers illuminate disease biology and treatment prospects. Progressing towards individualized efficient therapies is imminent, alleviating lung cancer's burden through ongoing research, omics integration, and technological strides.
Collapse
Affiliation(s)
- Shriyansh Srivastava
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi 110017, India; Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India
| | - Nandani Jayaswal
- Accurate College of Pharmacy, 49, Knowledge Park-III, Greater Noida, UP, India
| | - Sachin Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi 110017, India
| | - Pramod Kumar Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Sahibzada Ajit Singh Nagar, Punjab, India.
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; Medicinal and Aromatic Plants Research Institute, National Center for Research, P.O. Box: 2424, Khartoum 11111, Sudan
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; Center for Global Health Research, Saveetha Medical College, and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India.
| | - Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan, Saudi Arabia
| | - Khalid Zoghebi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan, Saudi Arabia
| | - Hassan A Alhazmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan, Saudi Arabia
| |
Collapse
|
44
|
Zhao Z, Chen Y, Sun T, Jiang C. Nanomaterials for brain metastasis. J Control Release 2024; 365:833-847. [PMID: 38065414 DOI: 10.1016/j.jconrel.2023.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/21/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
Tumor metastasis is a significant contributor to the mortality of cancer patients. Specifically, current conventional treatments are unable to achieve complete remission of brain metastasis. This is due to the unique pathological environment of brain metastasis, which differs significantly from peripheral metastasis. Brain metastasis is characterized by high tumor mutation rates and a complex microenvironment with immunosuppression. Additionally, the presence of blood-brain barrier (BBB)/blood tumor barrier (BTB) restricts drug leakage into the brain. Therefore, it is crucial to take account of the specific characteristics of brain metastasis when developing new therapeutic strategies. Nanomaterials offer promising opportunities for targeted therapies in treating brain metastasis. They can be tailored and customized based on specific pathological features and incorporate various treatment approaches, which makes them advantageous in advancing therapeutic strategies for brain metastasis. This review provides an overview of current clinical treatment options for patients with brain metastasis. It also explores the roles and changes that different cells within the complex microenvironment play during tumor spread. Furthermore, it highlights the use of nanomaterials in current brain treatment approaches.
Collapse
Affiliation(s)
- Zhenhao Zhao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yun Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
45
|
Salmani-Javan E, Farhoudi Sefidan Jadid M, Zarghami N. Recent advances in molecular targeted therapy of lung cancer: Possible application in translation medicine. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:122-133. [PMID: 38234663 PMCID: PMC10790298 DOI: 10.22038/ijbms.2023.72407.15749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/23/2023] [Indexed: 01/19/2024]
Abstract
Lung cancer is one of the leading causes of death among all cancer deaths. This cancer is classified into two different histological subtypes: non-small cell lung cancer (NSCLC), which is the most common subtype, and small cell lung cancer (SCLC), which is the most aggressive subtype. Understanding the molecular characteristics of lung cancer has expanded our knowledge of the cellular origins and molecular pathways affected by each of these subtypes and has contributed to the development of new therapies. Traditional treatments for lung cancer include surgery, chemotherapy, and radiotherapy. Advances in understanding the nature and specificity of lung cancer have led to the development of immunotherapy, which is the newest and most specialized treatment in the treatment of lung cancer. Each of these treatments has advantages and disadvantages and causes side effects. Today, combination therapy for lung cancer reduces side effects and increases the speed of recovery. Despite the significant progress that has been made in the treatment of lung cancer in the last decade, further research into new drugs and combination therapies is needed to extend the clinical benefits and improve outcomes in lung cancer. In this review article, we discussed common lung cancer treatments and their combinations from the most advanced to the newest.
Collapse
Affiliation(s)
- Elnaz Salmani-Javan
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Farhoudi Sefidan Jadid
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| |
Collapse
|
46
|
Pavlovic D, Niciforovic D, Markovic M, Papic D. Cancer-Associated Thrombosis: Epidemiology, Pathophysiological Mechanisms, Treatment, and Risk Assessment. Clin Med Insights Oncol 2023; 17:11795549231220297. [PMID: 38152726 PMCID: PMC10752082 DOI: 10.1177/11795549231220297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/28/2023] [Indexed: 12/29/2023] Open
Abstract
Cancer patients represent a growing population with drastically difficult care and a lowered quality of life, especially due to the heightened risk of vast complications. Thus, it is well established so far that one of the most prominent complications in individuals with cancer is venous thromboembolism. Since there are various improved methods for screening and diagnosing cancer and its complications, the incidence of cancer-associated thrombosis has been on the rise in recent years. Therefore, the high mortality and morbidity rates among these patients are not a surprise. Consequently, there is an excruciating need for understanding the mechanisms behind this complex process, as well as the imperative for adequate analysis and application of the most suitable steps for cancer-associated thrombosis prevention. There are various and numerous mechanisms offering potential answers to cancer-associated thrombosis, some of which have already been elucidated in various preclinical and clinical scenarios, yet further and more elaborate studies are crucial to understanding and preventing this complex and harsh clinical entity. This article elaborates on the growing incidence, mortality, morbidity, and risk factors of cancer-associated thrombosis while emphasizing the pathophysiological mechanisms in the light of various types of cancer in patients and summarizes the most novel therapy and prevention guidelines recommendations.
Collapse
Affiliation(s)
- Dragica Pavlovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Danijela Niciforovic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Marina Markovic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragana Papic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
47
|
Noguchi K, Ikawa Y. Strategy for Pediatric Patients with Relapsed or Refractory Anaplastic Lymphoma Kinase-Positive Anaplastic Large Cell Lymphoma: A Review. Cancers (Basel) 2023; 15:5733. [PMID: 38136278 PMCID: PMC10741612 DOI: 10.3390/cancers15245733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/10/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Anaplastic lymphoma kinase (ALK)-positive anaplastic large cell lymphoma (ALCL) is an aggressive T-cell lymphoma characterized by large T-cells with strong CD30 and ALK expression. Although conventional chemotherapy is effective in most patients, approximately 30% experience a relapse or refractory disease and have a poor prognosis. Several risk factors associated with poor prognosis have been identified in pediatric ALK-positive ALCL. These include morphological patterns with the small cell variant or lymphohistiocytic variant, leukemic presentation, the presence of minimal disseminated disease, or involvement of the central nervous system. Relapsed or refractory ALK-positive ALCL is often resistant to conventional chemotherapy; therefore, salvage therapy is required. In recent years, targeted therapies such as ALK inhibitors and brentuximab vedotin (BV) have been developed. ALK inhibitors block the continuous activation of ALK kinase, a driver mutation that leads to cell proliferation in ALK-positive ALCL. Additionally, BV is an antibody-drug conjugate that targets CD30-positive cells. Both ALK inhibitors and BV have displayed dramatic effects in chemoresistant ALK-positive ALCL. Weekly vinblastine treatment and hematopoietic stem cell transplantation have also been reported to be effective therapies. This article reviews pediatric ALK-positive ALCL, focusing on risk factors and treatment strategies for pediatric patients with relapsed or refractory ALK-positive ALCL.
Collapse
Affiliation(s)
| | - Yasuhiro Ikawa
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan;
| |
Collapse
|
48
|
Hansen KH, Johansen JS, Urbanska EM, Meldgaard P, Hjorth-Hansen P, Kristiansen C, Stelmach M, Santoni-Rugiu E, Ulhøi MP, Dydensborg AB, Dünweber C, Andersen JL. Clinical outcomes of ALK+ non-small cell lung cancer in Denmark. Acta Oncol 2023; 62:1775-1783. [PMID: 37815923 DOI: 10.1080/0284186x.2023.2263153] [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: 07/19/2023] [Accepted: 09/21/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Real-world clinical outcomes of anaplastic lymphoma kinase positive (ALK+) non-small cell lung cancer (NSCLC) patients vary. This study aimed to investigate the treatment and clinical outcomes of all ALK+ NSCLC patients in Denmark in the period 2011-2018, regardless of disease stage. MATERIALS AND METHODS A national pathology database with complete coverage was used to identify ALK+ NSCLC patients diagnosed between 2011 and 2018. Clinical data were obtained through retrospective chart reviews. Overall survival (OS) and duration of treatment (DOT) were analyzed using Kaplan-Meier methodologies. RESULTS A total of 209 ALK+ NSCLC patients were included. The cohort had a slight overrepresentation of female patients (56.5%) with a mean age of 61.6 years. Most patients were adenocarcinoma cases (97%) and presented with an ECOG performance status of 0-1 (79%). Stage IIIb-IVb patients comprised 70% of the cohort. The use of ALK-tyrosine kinase inhibitors (TKIs) as first-line treatment increased over time, with the 1st generation ALK-TKI crizotinib being the predominant treatment in the 1st line. In 1st line treatment, 2nd generation ALK-TKIs had a median DOT more than twice the median DOT of crizotinib (25.1 and 9.1 months, respectively). The median OS for the entire cohort was 44.0 months. Patients with stage I-IIIA disease had a median OS that had not been reached, while those with stage IIIb-IVb disease had a median OS of 31.8 months. Patients with stage IIIb-IVb disease receiving an ALK-TKI as 1st line treatment had a median OS of 42.5 months with immature follow-up. Brain metastases at diagnosis or choice of 1st line treatment did not statistically significantly impact OS. CONCLUSION This study gives insights into the treatment and outcome of ALK+ NSCLC patients in Denmark and provides a real-world confirmation of the superior disease control provided by 2nd generation ALK-TKIs as compared to the 1st generation ALK-TKI crizotinib.
Collapse
Affiliation(s)
| | | | - Edyta Maria Urbanska
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Peter Meldgaard
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Charlotte Kristiansen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | | | - Eric Santoni-Rugiu
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
49
|
Fischer M, Luck M, Werle M, Vogel A, Bashawat M, Ludwig K, Scheidt HA, Müller P. The small-molecule kinase inhibitor ceritinib, unlike imatinib, causes a significant disturbance of lipid membrane integrity: A combined experimental and MD study. Chem Phys Lipids 2023; 257:105351. [PMID: 37863350 DOI: 10.1016/j.chemphyslip.2023.105351] [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: 07/11/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Ceritinib and imatinib are small-molecule protein kinase inhibitors which are applied as therapeutic agents against various diseases. The fundamentals of their clinical use, i.e. their pharmacokinetics as well as the mechanisms of the inhibition of the respective kinases, are relatively well studied. However, the interaction of the drugs with membranes, which can be a possible cause of side effects, has hardly been investigated so far. Therefore, we have characterized the interaction of both drugs with lipid membranes consisting of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) in the absence and in the presence of cholesterol. For determining the membrane impact of both drugs on a molecular level, different experimental (NMR, ESR, fluorescence) and theoretical (MD simulations) approaches were applied. The data show that ceritinib, in contrast to imatinib, interacts more effectively with membranes significantly affecting various physico-chemical membrane parameters like membrane order and transmembrane permeation of polar solutes. The pronounced membrane impact of ceritinib can be explained by a strong affinity of the drug towards POPC which competes with the POPC-cholesterol interaction by that attenuating the ordering effect of cholesterol. The data are relevant for understanding putative toxic and cytotoxic side effects of these drugs such as the triggering of cell lysis or apoptosis.
Collapse
Affiliation(s)
- Markus Fischer
- Leipzig University, Institute for Medical Physics and Biophysics, Härtelstr. 16-18, D-04107 Leipzig, Germany
| | - Meike Luck
- Humboldt University Berlin, Department of Biology, Invalidenstr. 42, D-10115 Berlin, Germany
| | - Max Werle
- Humboldt University Berlin, Department of Biology, Invalidenstr. 42, D-10115 Berlin, Germany
| | - Alexander Vogel
- Leipzig University, Institute for Medical Physics and Biophysics, Härtelstr. 16-18, D-04107 Leipzig, Germany
| | - Mohammad Bashawat
- Humboldt University Berlin, Department of Biology, Invalidenstr. 42, D-10115 Berlin, Germany
| | - Kai Ludwig
- Freie Universität Berlin, Research Center for Electron Microscopy and Core Facility BioSupraMol, Institute of Chemistry and Biochemistry, Fabeckstr. 36a, D-14195 Berlin, Germany
| | - Holger A Scheidt
- Leipzig University, Institute for Medical Physics and Biophysics, Härtelstr. 16-18, D-04107 Leipzig, Germany.
| | - Peter Müller
- Humboldt University Berlin, Department of Biology, Invalidenstr. 42, D-10115 Berlin, Germany.
| |
Collapse
|
50
|
Gao H, Zhang JY, Zhao LJ, Guo YY. Synthesis and clinical application of small-molecule inhibitors and PROTACs of anaplastic lymphoma kinase. Bioorg Chem 2023; 140:106807. [PMID: 37651895 DOI: 10.1016/j.bioorg.2023.106807] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/13/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023]
Abstract
Pharmacological interventions that specifically target protein products of oncogenes in tumors have surfaced as a propitious therapeutic approach. Among infrequent genetic alterations, rearrangements of the anaplastic lymphoma kinase (ALK) gene, typically involving a chromosome 2 inversion that culminates in a fusion with the echinoderm microtubule-associated protein like 4 (EML4), lead to anomalous expression and activation of ALK. The inhibition of autophosphorylation and subsequent blockade of signal transduction by ALK tyrosine kinase inhibitors (TKIs) has been observed to elicit anti-tumor effects. Currently, four generations of ALK-positive targeted drugs have been investigated, providing a promising outlook for patients. The aim of this review is to furnish a comprehensive survey of the synthesis and clinical application of prototypical small-molecule ALK inhibitors in both preclinical and clinical phases, offering guidance for further development of ALK inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Hua Gao
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jing-Yi Zhang
- The Rogel Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States; College of Chemistry and Chemical Engineering, Zhengzhou Normal University 450044, China.
| | - Li-Jie Zhao
- The Rogel Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Yuan-Yuan Guo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|