1
|
Liu QG, Wu J, Wang ZY, Chen BB, Du YF, Niu JB, Song J, Zhang SY. ALK-based dual inhibitors: Focus on recent development for non-small cell lung cancer therapy. Eur J Med Chem 2025; 291:117646. [PMID: 40262298 DOI: 10.1016/j.ejmech.2025.117646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
As a prevalent oncogenic driver gene in non-small cell lung cancer (NSCLC), ALK represents a crucial and efficacious therapeutic target. To date, seven ALK inhibitors have been approved for ALK fusion-positive NSCLC, with several others undergoing clinical trials. These therapies demonstrate significant efficacy in ALK fusion-positive NSCLC patients. However, acquired resistance mechanisms, including ALK kinase domain mutations, ALK gene amplification, and bypass pathway activation, significantly compromise the efficacy of targeted therapy in ALK fusion-positive NSCLC. Therefore, the discovery of novel ALK inhibitors and the development of related treatment strategies remain critical. Compared to the combination therapy strategy based on ALK inhibitors, dual-target inhibitors (targeting two distinct pathways within a single molecule) may reduce systemic toxicity and mitigate resistance mechanisms in cancer treatment. Notably, recent years have witnessed remarkable progress in dual-target ALK inhibitor development for NSCLC. Consequently, this review aims to summarize the advancements achieved through dual ALK-based inhibitors in NSCLC therapy, analyze their rational design and structure-activity relationships, and provide perspectives for overcoming resistance through next-generation inhibitors and innovative therapeutic approaches.
Collapse
Affiliation(s)
- Qiu-Ge Liu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Ji Wu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Zi-Yue Wang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Bing-Bing Chen
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yi-Fei Du
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jin-Bo Niu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
2
|
Xu J, Zhang W, Xie F, Wang C, Cheng F, Rao R, Chen Y, Zhang L, Wen W, Zhao Z, Yuan J, Zheng Y, Yu Z. Prognostic significance of ALK high expression in SCLC: a 9-year cohort analysis. Front Oncol 2025; 15:1530339. [PMID: 40201354 PMCID: PMC11975910 DOI: 10.3389/fonc.2025.1530339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose The aim of this study was to investigate the prognostic value of the abnormal expression of anaplastic lymphoma kinase (ALK) protein in patients with small cell lung cancer (SCLC) based on 9-year data from our center. Methods A retrospective cohort study was conducted to assess the clinical outcomes of patients with ALK-positive SCLC diagnosed in our hospital over the past 9 years. We used public databases to analyze the expression of ALK in pan-cancer and its prognostic value and analyzed the correlation between ALK and SCLC prognosis-related genes. Results A total of 685 patients diagnosed with SCLC underwent ALK testing, and 59 patients were identified to have abnormal expression of the ALK protein, with 10 cases showing strong expression, 14 cases displaying moderate expression, and 35 cases exhibiting weak expression. The median age of the ALK-positive cohort was 64 years (range: 58-70 years), 91.5% (54/59) were male, 61.0% (36/59) were smokers, and the median overall survival (mOS) was 7.0 months (95% CI: 4.5-9.5 months). Within this cohort, the mOS for the ALK (+) subgroup was 4.0 months (95% CI: 2.9-5.1 months), the mOS for the ALK (++) subgroup was 10.0 months (95% CI: 4.9-15.1 months), and the mOS for the ALK (+++) subgroup was 12.0 months (95% CI: 7.4-16.6 months). Kaplan-Meier revealed that the mOS of the ALKLow group was significantly worse than that of the ALKHigh group [mOS: 4.0 months (95% CI: 2.9-5.1 months) versus 11.0 months (95% CI: 8.3-13.7 months), p = 0.009]. Following covariate adjustment using a Cox regression model, it was indicated that the level of abnormal expression of the ALK protein was an independent prognostic factor for patients with SCLC (HR: 0.486, 95% CI: 0.271-0.871, p = 0.015). Conclusion The prognosis for patients with SCLC with strong abnormal expression of the ALK protein was significantly better than those with weak expression.
Collapse
Affiliation(s)
- Jinhe Xu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wenting Zhang
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Feilai Xie
- Pathology Department, Dongfang Hospital of Xiamen University, Fuzhou General Hospital of Fujian Medical University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Chenxi Wang
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Feng Cheng
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Ruiying Rao
- Department of Respiratory and Critical Care Medicine, Fuzong Teaching Hospital, Fujian University of Traditional Chinese Medicine (900 Hospital), Fuzhou, China
| | - Ying Chen
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Lei Zhang
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Wen Wen
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Zhongquan Zhao
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Jialing Yuan
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Yuqin Zheng
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Zongyang Yu
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| |
Collapse
|
3
|
Ye W, Ou WB. Genomic landscaping of receptor tyrosine kinase ALK with highly frequent rearrangements in cancers. IUBMB Life 2025; 77:e70003. [PMID: 39917830 DOI: 10.1002/iub.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/16/2025] [Indexed: 02/09/2025]
Abstract
Anaplastic lymphoma kinase (ALK) fusion tyrosine kinases (TKs) are commonly found in various cancers and are considered as promising targets for therapy due to their intricate biological processes. However, the reasons for the higher frequency of ALKs fusion compared to other TKs are not well elucidated. Physicochemical properties, secondary and tertiary structures, and phylogenetic trees, along with protein sequence alignments of receptor tyrosine kinases (RTKs) and ALK fused partner genes, were examined using the resources provided by the National Center for Biotechnology Information (NCBI) and the Catalogue of Somatic Mutations in Cancer (COSMIC). Sequence alignments were performed to identify common sequences between partner genes and search for common breakpoints within the COSMIC database. ALK is a large, unstable, acidic protein with similarly conservation among RTKs. ALK fusion partners are mostly acidic, unstable proteins, mostly consisting of α-helices and random coil. However, EML4 and NPM1 are the most frequently occurring partner genes and have their own unique structural characteristics. By functional domain analysis, we found that the functions of the first half of the ALK partner gene (the part fused to ALK) are mostly focused on signaling. ALK is identified as a large hydrophilic protein,exhibits a higher proportion of random coils. Compared to other RTKs, ALK has fewer structural domains (PTKC_ALK_LTK domain). Pairwise comparison with fusion partner genes revealed a conserved sequence predicted to have structural stability and act as a common binding site for nucleases. Exon 20 of ALK is a fusion frequent site according to COSMIC database analysis. The structural instability of ALK and partner genes, coupled with the inherent variability of breakpoint sequences, leads to the formation of potent kinase-activated oncogenes, which play a critical role in tumorigenesis. While the occurrence of ALK fusions with partner genes is random, specific combinations lead to the generation of oncogenes.
Collapse
Affiliation(s)
- Wei Ye
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Wen-Bin Ou
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
4
|
Hamidi S, Ning MS, Phan J, Zafereo ME, Gule-Monroe MK, Dadu R. Recurrent Poorly Differentiated Thyroid Cancer Successfully Treated With Radiation and Immunotherapy. JCEM CASE REPORTS 2025; 3:luaf015. [PMID: 39866918 PMCID: PMC11758140 DOI: 10.1210/jcemcr/luaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Indexed: 01/28/2025]
Abstract
A 65-year-old patient presented with recurrent, locally advanced poorly differentiated thyroid cancer despite 2 neck surgeries, and with newly diagnosed brain and skull base metastases. He was treated with palliative stereotactic radiosurgery to the brain and skull base lesions. Thereafter, as no targetable genetic alteration was identified and antiangiogenic multikinase inhibitors were deemed at high risk of hemorrhagic complications, off-label systemic therapies were considered. The mechanistic target of rapamycin (mTOR) inhibitor everolimus could not be obtained due to lack of insurance coverage, so the patient was treated with single-agent pembrolizumab. He showed an initial remarkable response, but unfortunately had disease progression in the neck and upper mediastinum after 1 year of therapy. At that time, he was treated with external beam radiotherapy, with concomitant pembrolizumab. He was then found to have an CTSB::ALK fusion, which has previously been described in 2 cases of thyroid cancer. However, as he showed a positive response to radiation with pembrolizumab, he continued single-agent immune checkpoint inhibition and had a persistent marked response almost a year after completing radiation. The patient was then followed at an outside institution and was transitioned to hospice at time of progression per his preference. He died 4 years after his initial diagnosis.
Collapse
Affiliation(s)
- Sarah Hamidi
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer, Houston, TX 77030, USA
| | - Matthew S Ning
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer, Houston, TX 77030, USA
| | - Jack Phan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer, Houston, TX 77030, USA
| | - Mark E Zafereo
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer, Houston, TX 77030, USA
| | - Maria K Gule-Monroe
- Department of Neuroradiology, The University of Texas MD Anderson Cancer, Houston, TX 77030, USA
| | - Ramona Dadu
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer, Houston, TX 77030, USA
| |
Collapse
|
5
|
Sorokin M, Lyadov V, Suntsova M, Garipov M, Semenova A, Popova N, Guguchkin E, Heydarov R, Zolotovskaia M, Zhao X, Yan Q, Wang Y, Karpulevich E, Buzdin A. Detection of fusion events by RNA sequencing in FFPE versus freshly frozen colorectal cancer tissue samples. Front Mol Biosci 2025; 11:1448792. [PMID: 39906487 PMCID: PMC11791353 DOI: 10.3389/fmolb.2024.1448792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/20/2024] [Indexed: 02/06/2025] Open
Abstract
Gene fusion events result in chimeric proteins that are frequently found in human cancers. Specific targeted therapies are available for several types of cancer fusions including receptor tyrosine kinase gene moieties. RNA sequencing (RNAseq) can directly be used for detection of gene rearrangements in a single test, along with multiple additional biomarkers. However, tumor biosamples are usually formalin-fixed paraffin-embedded (FFPE) tissue blocks where RNA is heavily degraded, which in theory may result in decreased efficiency of fusion detection. Here, for the first time, we compared the efficacy of gene fusion detection by RNAseq for matched pairs of freshly frozen in RNA stabilizing solution (FF) and FFPE tumor tissue samples obtained from 29 human colorectal cancer patients. We detected no statistically significant difference in the number of chimeric transcripts in FFPE and FF RNAseq profiles. The known fusion KANSL1-ARL17A/B occurred with a high frequency in 69% of the patients. We also detected 93 new fusion genes not mentioned in the literature or listed in the ChimerSeq database. Among them, 11 were found in two or more patients, suggesting their potential role in carcinogenesis. Most of the fusions detected most probably represented read-through, microdeletion or local duplication events. Finally, in one patient, we detected a potentially clinically actionable in-frame fusion of LRRFIP2 and ALK genes not previously described in colorectal cancer with an intact tyrosine kinase domain that can be potentially targeted by ALK inhibitors.
Collapse
Affiliation(s)
- Maxim Sorokin
- OmicsWay Corp., Covina, CA, United States
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Moscow Center for Advanced Studies, Moscow, Russia
| | - Vladimir Lyadov
- Moscow State Budgetary Healthcare Institution “Moscow City Oncological Hospital N1, Moscow Healthcare Department”, Moscow, Russia
- Federal State Budgetary Educational Institution of Further Professional Education “Russian Medical Academy of Continuous Professional Education” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
- Novokuznetsk State Institute for Advanced Training of Physicians – Branch of RMACPE, Novokuznetsk, Russia
| | - Maria Suntsova
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Marat Garipov
- Moscow State Budgetary Healthcare Institution “Moscow City Oncological Hospital N1, Moscow Healthcare Department”, Moscow, Russia
| | - Anna Semenova
- Moscow State Budgetary Healthcare Institution “Moscow City Oncological Hospital N1, Moscow Healthcare Department”, Moscow, Russia
| | - Natalia Popova
- Moscow State Budgetary Healthcare Institution “Moscow City Oncological Hospital N1, Moscow Healthcare Department”, Moscow, Russia
| | | | - Rustam Heydarov
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Xiaowen Zhao
- Core lab, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Qing Yan
- Core lab, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Ye Wang
- Core lab, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | | | - Anton Buzdin
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium
- Moscow Center for Advanced Studies, Moscow, Russia
- Group for Genomic Regulation of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
6
|
Adashek JJ, Munoz JL, Kurzrock R. If it is a solid tumor target, then it may be a hematologic cancer target: Bridging the great divide. MED 2025; 6:100550. [PMID: 39689708 PMCID: PMC11725447 DOI: 10.1016/j.medj.2024.11.003] [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: 04/26/2024] [Revised: 10/01/2024] [Accepted: 11/01/2024] [Indexed: 12/19/2024]
Abstract
Tumor-agnostic US Food and Drug Administration approvals are transforming oncology. They include larotrectinib/entrectinib/repotrectinib (NTRK fusions), selpercatinib (RET fusions), dabrafenib/trametinib (BRAFV600E mutations), pembrolizumab/dostarlimab (microsatellite instability), pembrolizumab (high tumor mutational burden), and trastuzumab deruxtecan (HER2 3+ expression) (all solid cancers). Pemigatinib is approved for FGFR1-rearranged myeloid/lymphoid neoplasms. The genomically driven tissue-agnostic approach has a strong biological rationale (cancer is a disease of the genome), yields remarkably high response rates, and provides drug access to patients with an unmet need (rare/ultra-rare malignancies). Despite the solid tumor focus, both solid and hematologic cancers can harbor identical driver molecular abnormalities and respond to cognate therapies. For example, BRAFV600E and IDH1/2 mutations; ALK, FGFR, and NTRK fusions; PD-L1 amplification; and CD70 antigens are druggable in both solid and blood malignancies by gene-/immune-targeted therapies/chimeric antigen receptor T cells. Future biomarker-based tissue-agnostic basket studies/approvals should bridge the great divide and include both solid and hematologic cancers.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Javier L Munoz
- Department of Hematology, Mayo Clinic Arizona, Phoenix, AZ, USA.
| | - Razelle Kurzrock
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA; WIN Consortium, Paris, France; University of Nebraska, Omaha, NE, USA.
| |
Collapse
|
7
|
Hiken J, Earls J, Flanagan KC, Wellinghoff RL, Ponder M, Messina DN, Glasscock JI, Duncavage EJ. Analytical performance of OncoPrism-HNSCC, an RNA-based assay to inform immune checkpoint inhibitor treatment decisions for recurrent/metastatic head and neck squamous cell carcinoma. BMC Cancer 2025; 25:21. [PMID: 39773366 PMCID: PMC11705923 DOI: 10.1186/s12885-024-13362-8] [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: 05/29/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND While immune checkpoint inhibitor (ICI) therapies can significantly improve outcomes for patients with recurrent/metastatic head and neck squamous cell carcinoma (RM-HNSCC), only about 15-20% benefit from such treatments. Clinical tests that guide the use of ICIs are therefore critically needed. OncoPrism-HNSCC was developed to address this need. The assay combines next generation RNA sequencing-based immunomodulatory gene expression signatures with machine learning algorithms to generate an OncoPrism score that classifies patients as having low, medium, or high likelihood of disease control in response to ICI treatment. Also, OncoPrism-HNSCC leverages the same FFPE patient tumor RNA used for ICI response prediction to identify rare cases where oncogenic rearrangements in NTRK1/2/3 or ALK genes may occur, and which may indicate the use of potentially highly effective targeted therapies. The clinical performance of OncoPrism-HNSCC has been validated. Here, we report its analytical performance in the presence of potentially confounding sources of variation. METHODS The assay's analytical sensitivity was assessed by varying RNA input quantity and quality, observing the effect on ICI response prediction scores. Analytical specificity was tested by spiking increasing percentages of genomic DNA into input RNA. Intra-assay and inter-assay precision were evaluated, and the analytical sensitivity, specificity, and precision of gene fusion detection were assessed. Concordance with orthogonal methods of gene fusion detection was tested on 67 FFPE clinical samples. RESULTS Varying RNA inputs as low as four-fold below the nominal input amount had little effect on ICI response prediction scores. RNA quality levels below the test threshold had no significant effect. Genomic DNA spike-ins up to 30% had only a small effect on scores. The pooled standard deviation for multiple operators, reagent lots, batches, and sequencers yielded an overall variance represented by just 0.87% of the score range of the test (0-100). NTRK and ALK gene fusion detection was 100% concordant with orthogonal methods. CONCLUSIONS Robust and reliable analytical performance of the OncoPrism-HNSCC assay supports its clinical use, even in the presence of variation typically encountered in the laboratory setting.
Collapse
Affiliation(s)
- Jeffrey Hiken
- Cofactor Genomics, Inc, 4044 Clayton Ave, St. Louis, MO, 63110, USA
| | - Jon Earls
- Cofactor Genomics, Inc, 4044 Clayton Ave, St. Louis, MO, 63110, USA
| | - Kevin C Flanagan
- Cofactor Genomics, Inc, 4044 Clayton Ave, St. Louis, MO, 63110, USA
| | | | - Michelle Ponder
- Cofactor Genomics, Inc, 4044 Clayton Ave, St. Louis, MO, 63110, USA
| | - David N Messina
- Cofactor Genomics, Inc, 4044 Clayton Ave, St. Louis, MO, 63110, USA
| | | | - Eric J Duncavage
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
8
|
Li Q, Cai T, Zheng X, Zhang S, Li C, Tang H, Yu Z, Zhou J. EML4-ALK-Positive Ovarian Cancer With Intracranial Metastasis Responds to Lorlatinib: A Case Report and Literature Review. Clin Case Rep 2025; 13:e70043. [PMID: 39780907 PMCID: PMC11707256 DOI: 10.1002/ccr3.70043] [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: 07/04/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
We report a case showing that lorlatinib is effective in treating EML4-ALK-positive low-grade serous ovarian cancer (LGSO) with intracranial metastasis. This may be the first clinical evidence of LGSO benefit from ALK inhibitors, to provide evidence for the use of ALK inhibitors in more ovarian cancer patients with EML4-ALK fusion and promoting new ideas for the study of EML4-ALK targets in ovarian cancer.
Collapse
Affiliation(s)
- Qiongqian Li
- Department of OncologyGuangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese MedicineGuangxiChina
| | - Tongze Cai
- Department of OncologyGuangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese MedicineGuangxiChina
| | - Xiaoming Zheng
- Department of OncologyGuangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese MedicineGuangxiChina
| | - Shunrong Zhang
- Department of OncologyGuangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese MedicineGuangxiChina
| | - Chanjuan Li
- Department of OncologyGuangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese MedicineGuangxiChina
| | - Huang Tang
- Department of OncologyGuangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese MedicineGuangxiChina
| | - Zhiyong Yu
- Department of OncologyGuangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese MedicineGuangxiChina
| | - Jianlong Zhou
- Department of OncologyGuangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese MedicineGuangxiChina
| |
Collapse
|
9
|
Mun J, Lim B. The hallmarks of tissue-agnostic therapies and strategies for early anticancer drug discovery. Drug Discov Today 2024; 29:104203. [PMID: 39389456 DOI: 10.1016/j.drudis.2024.104203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Recently, precision medicine has enabled biomarker-driven treatment to be administered in a pan-tumor setting, so-called tissue-agnostic therapy. This represents a paradigm shift in cancer care from conventional cancer type-specific to biomarker-centric care, where patients are selected for this type of therapy based on molecular aberrations that occur across cancer types, regardless of tumor site or histology. This approach is particularly important because it offers new treatment options for patients with rare cancers or for whom there are no adequate treatments. Therefore drug development with the goal of tissue agnosticism can accelerate drug approval and address unmet medical needs. Herein, we discuss our opinions on the hallmarks of tissue-agnostic treatments and propose strategies for early drug discovery based on these hallmarks.
Collapse
Affiliation(s)
- Jihyeob Mun
- Center for Supercomputing Applications, Division of National Supercomputing R&D, Korea Institute of Science and Technology Information (KISTI), Daejeon, Republic of Korea
| | - Byungho Lim
- Data Convergence Drug Research Center, Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, University of Science and Technology (UST), Daejeon, Republic of Korea.
| |
Collapse
|
10
|
AlDoughaim M, AlSuhebany N, AlZahrani M, AlQahtani T, AlGhamdi S, Badreldin H, Al Alshaykh H. Cancer Biomarkers and Precision Oncology: A Review of Recent Trends and Innovations. Clin Med Insights Oncol 2024; 18:11795549241298541. [PMID: 39559827 PMCID: PMC11571259 DOI: 10.1177/11795549241298541] [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: 07/08/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024] Open
Abstract
The discovery of cancer-specific biomarkers has resulted in major advancements in the field of cancer diagnostics and therapeutics, therefore significantly lowering cancer-related morbidity and mortality. Cancer biomarkers can be generally classified as prognostic biomarkers that predict specific disease outcomes and predictive biomarkers that predict disease response to targeted therapeutic interventions. As research in the area of predictive biomarkers continues to grow, precision medicine becomes far more integrated in cancer treatment. This article presents a general overview on the most recent advancements in the area of cancer biomarkers, immunotherapy, artificial intelligence, and pharmacogenomics of the Middle East.
Collapse
Affiliation(s)
- Maha AlDoughaim
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Nada AlSuhebany
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Mohammed AlZahrani
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Tariq AlQahtani
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Sahar AlGhamdi
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Hisham Badreldin
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Hana Al Alshaykh
- Pharmaceutical Care Devision, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Zakharova G, Suntsova M, Rabushko E, Mohammad T, Drobyshev A, Seryakov A, Poddubskaya E, Moisseev A, Smirnova A, Sorokin M, Tkachev V, Simonov A, Guguchkin E, Karpulevich E, Buzdin A. A New Approach of Detecting ALK Fusion Oncogenes by RNA Sequencing Exon Coverage Analysis. Cancers (Basel) 2024; 16:3851. [PMID: 39594806 PMCID: PMC11592821 DOI: 10.3390/cancers16223851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND In clinical practice, various methods are used to identify ALK gene rearrangements in tumor samples, ranging from "classic" techniques, such as IHC, FISH, and RT-qPCR, to more advanced highly multiplexed approaches, such as NanoString technology and NGS panels. Each of these methods has its own advantages and disadvantages, but they share the drawback of detecting only a restricted (although sometimes quite extensive) set of preselected biomarkers. At the same time, whole transcriptome sequencing (WTS, RNAseq) can, in principle, be used to detect gene fusions while simultaneously analyzing an incomparably wide range of tumor characteristics. However, WTS is not widely used in practice due to purely analytical limitations and the high complexity of bioinformatic analysis, which requires considerable expertise. In particular, methods to detect gene fusions in RNAseq data rely on the identification of chimeric reads. However, the typically low number of true fusion reads in RNAseq limits its sensitivity. In a previous study, we observed asymmetry in the RNAseq exon coverage of the 3' partners of some fusion transcripts. In this study, we conducted a comprehensive evaluation of the accuracy of ALK fusion detection through an analysis of differences in the coverage of its tyrosine kinase exons. METHODS A total of 906 human cancer biosamples were subjected to analysis using experimental RNAseq data, with the objective of determining the extent of asymmetry in ALK coverage. A total of 50 samples were analyzed, comprising 13 samples with predicted ALK fusions and 37 samples without predicted ALK fusions. These samples were assessed by targeted sequencing with two NGS panels that were specifically designed to detect fusion transcripts (the TruSight RNA Fusion Panel and the OncoFu Elite panel). RESULTS ALK fusions were confirmed in 11 out of the 13 predicted cases, with an overall accuracy of 96% (sensitivity 100%, specificity 94.9%). Two discordant cases exhibited low ALK coverage depth, which could be addressed algorithmically to enhance the accuracy of the results. It was also important to consider read strand specificity due to the presence of antisense transcripts involving parts of ALK. In a limited patient sample undergoing ALK-targeted therapy, the algorithm successfully predicted treatment efficacy. CONCLUSIONS RNAseq exon coverage analysis can effectively detect ALK rearrangements.
Collapse
Affiliation(s)
- Galina Zakharova
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
| | - Maria Suntsova
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Endocrinology Research Center, 117292 Moscow, Russia;
| | - Elizaveta Rabushko
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
| | | | - Alexey Drobyshev
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
| | | | - Elena Poddubskaya
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Clinical Center Vitamed, 121309 Moscow, Russia
| | - Alexey Moisseev
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Oncobox LLC, 119991 Moscow, Russia;
| | - Anastasia Smirnova
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Oncobox LLC, 119991 Moscow, Russia;
| | - Maxim Sorokin
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Oncobox LLC, 119991 Moscow, Russia;
| | | | - Alexander Simonov
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
| | - Egor Guguchkin
- Institute for System Programming of RAS, 109004 Moscow, Russia; (E.G.); (E.K.)
| | - Evgeny Karpulevich
- Institute for System Programming of RAS, 109004 Moscow, Russia; (E.G.); (E.K.)
| | - Anton Buzdin
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| |
Collapse
|
12
|
Bhamidipati D, Schram AM. Emerging Tumor-Agnostic Molecular Targets. Mol Cancer Ther 2024; 23:1544-1554. [PMID: 39279103 PMCID: PMC11908425 DOI: 10.1158/1535-7163.mct-23-0725] [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: 04/24/2024] [Revised: 07/30/2024] [Accepted: 09/06/2024] [Indexed: 09/18/2024]
Abstract
Advances in tumor molecular profiling have uncovered shared genomic and proteomic alterations across tumor types that can be exploited therapeutically. A biomarker-driven, disease-agnostic approach to oncology drug development can maximize the reach of novel therapeutics. To date, eight drug-biomarker pairs have been approved for the treatment of patients with advanced solid tumors with specific molecular profiles. Emerging biomarkers with the potential for clinical actionability across tumor types include gene fusions involving NRG1, FGFR1/2/3, BRAF, and ALK and mutations in TP53 Y220C, KRAS G12C, FGFR2/3, and BRAF non-V600 (class II). We explore the growing evidence for clinical actionability of these biomarkers in patients with advanced solid tumors.
Collapse
Affiliation(s)
| | - Alison M. Schram
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
13
|
Suzuki Y, Tsubota S, Kadomatsu K, Sakamoto K. Identification of APBB1 as a substrate for anaplastic lymphoma kinase. J Biochem 2024; 176:395-403. [PMID: 39115278 PMCID: PMC11954158 DOI: 10.1093/jb/mvae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/15/2024] [Indexed: 11/05/2024] Open
Abstract
Anaplastic lymphoma kinase (ALK) is a well-known oncogene involved in various malignancies such as anaplastic large cell lymphoma, lung cancer and neuroblastoma. Several substrates for fused ALK have been identified and their biological functions have been described. However, the lack of a comprehensive identification of ALK substrates limits our understanding of the biological roles of receptor ALK. Thus, this study aimed to identify novel ALK substrates and characterize their biological functions. We screened the interactors of the kinase domain of receptor ALK using proximity-dependent biotin identification and identified 43 interactors. We narrowed down the candidates by evaluating whether these interactors were downstream of ALK in a neuroblastoma cell line, NB-1. Amongst these, we identified amyloid beta precursor protein-binding family B member 1 (APBB1) as an ALK downstream molecule involved in NB-1 cell viability. Finally, we assessed the kinase-substrate relationship between ALK and APBB1 and found that ALK phosphorylated multiple tyrosine residues in APBB1 both in-cell and in-tube assays, with tyrosine 269 as a major target. In conclusion, we successfully identified a new substrate for receptor ALK. Our results may help further elucidate the molecular mechanism of ALK downstream signalling in neuroblastoma.
Collapse
Affiliation(s)
- Yuji Suzuki
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
- Department of Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Shoma Tsubota
- Department of Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Kenji Kadomatsu
- Institute for Glyco-core Research, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8601, Japan
| | - Kazuma Sakamoto
- Department of Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
- Institute for Glyco-core Research, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8601, Japan
| |
Collapse
|
14
|
Dinić J, Jovanović Stojanov S, Dragoj M, Grozdanić M, Podolski-Renić A, Pešić M. Cancer Patient-Derived Cell-Based Models: Applications and Challenges in Functional Precision Medicine. Life (Basel) 2024; 14:1142. [PMID: 39337925 PMCID: PMC11433531 DOI: 10.3390/life14091142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/22/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
The field of oncology has witnessed remarkable progress in personalized cancer therapy. Functional precision medicine has emerged as a promising avenue for achieving superior treatment outcomes by integrating omics profiling and sensitivity testing of patient-derived cancer cells. This review paper provides an in-depth analysis of the evolution of cancer-directed drugs, resistance mechanisms, and the role of functional precision medicine platforms in revolutionizing individualized treatment strategies. Using two-dimensional (2D) and three-dimensional (3D) cell cultures, patient-derived xenograft (PDX) models, and advanced functional assays has significantly improved our understanding of tumor behavior and drug response. This progress will lead to identifying more effective treatments for more patients. Considering the limited eligibility of patients based on a genome-targeted approach for receiving targeted therapy, functional precision medicine provides unprecedented opportunities for customizing medical interventions according to individual patient traits and individual drug responses. This review delineates the current landscape, explores limitations, and presents future perspectives to inspire ongoing advancements in functional precision medicine for personalized cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11108 Belgrade, Serbia; (J.D.); (S.J.S.); (M.D.); (M.G.); (A.P.-R.)
| |
Collapse
|
15
|
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
|
16
|
Chang YH, Yu CH, Lu MY, Jou ST, Lin CY, Lin KH, Chang HH, Ni YL, Chou SW, Ko KY, Lin DT, Hsu WM, Chen HY, Yang YL. Higher tumor mutational burden is associated with inferior outcomes among pediatric patients with neuroblastoma. Pediatr Blood Cancer 2024; 71:e31176. [PMID: 38967585 DOI: 10.1002/pbc.31176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 05/18/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
INTRODUCTION Neuroblastoma is a pediatric malignancy with heterogeneous clinical outcomes. Our aim was to identify prognostic genetic markers for patients with neuroblastoma, who were treated with the Taiwan Pediatric Oncology Group (TPOG) neuroblastoma N2002 protocol, to improve risk stratification and inform treatment. METHODS Our analysis was based on 53 primary neuroblastoma specimens, diagnosed pre-chemotherapy, and 11 paired tumor relapse specimens. Deep sequencing of 113 target genes was performed using a custom panel. Multiplex ligation-dependent probe amplification was performed to identify clinical outcomes related to copy-number variations. RESULTS We identified 128 variations associated with survival, with the number of variations being higher in the relapse than that in the diagnostic specimen (p = .03). The risk of event and mortality was higher among patients with a tumor mutational burden ≥10 than that in patients with a lower burden (p < .0001). Multivariate analysis identified tumor mutational burden, MYCN amplification, and chromosome 3p deletion as significant prognostic factors, independent of age at diagnosis, sex, and tumor stage. The 5-year event-free survival and overall survival rate was lower among patients with high tumor burden than in patients with low tumor burden. Furthermore, there was no survival of patients with an ALK F1147L variation at 5 years after diagnosis. CONCLUSIONS Genome sequencing to determine the tumor mutational burden and ALK variations can improve the risk classification of neuroblastoma and inform treatment.
Collapse
Affiliation(s)
- Ya-Hsuan Chang
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan
| | - Chih-Hsiang Yu
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
- Departments of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Meng-Yao Lu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shiann-Tarng Jou
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Yu Lin
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
| | - Kai-Hsin Lin
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsiu-Hao Chang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Ling Ni
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shu-Wei Chou
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuan-Yin Ko
- Department of Nuclear Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Dong-Tsamn Lin
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Department of Surgery, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsuan-Yu Chen
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
| | - Yung-Li Yang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Laboratory Medicine and Medical Service, National Taiwan University Cancer Center, Taipei, Taiwan
| |
Collapse
|
17
|
Nikanjam M, Wells K, Kato S, Adashek JJ, Block S, Kurzrock R. Reverse repurposing: Potential utility of cancer drugs in nonmalignant illnesses. MED 2024; 5:689-717. [PMID: 38749442 PMCID: PMC11246816 DOI: 10.1016/j.medj.2024.04.008] [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: 03/01/2024] [Revised: 04/03/2024] [Accepted: 04/18/2024] [Indexed: 06/02/2024]
Abstract
Growth and immune process dysregulation can result in both cancer and nonmalignant disease (hereditary or acquired, with and without predisposition to malignancy). Moreover, perhaps unexpectedly, many nonmalignant illnesses harbor genomic alterations indistinguishable from druggable oncogenic drivers. Therefore, targeted compounds used successfully to treat cancer may have therapeutic potential for nonmalignant conditions harboring the same target. MEK, PI3K/AKT/mTOR, fibroblast growth factor receptor (FGFR), and NRG1/ERBB pathway genes have all been implicated in both cancer and noncancerous conditions, and several cognate antagonists, as well as Bruton's tyrosine kinase inhibitors, JAK inhibitors, and CD20-directed antibodies, have established or theoretical therapeutic potential to bridge cancer and benign diseases. Intriguingly, pharmacologically tractable cancer drivers characterize a wide spectrum of disorders without malignant potential, including but not limited to Alzheimer's disease and a variety of other neurodegenerative conditions, rheumatoid arthritis, achondroplastic dwarfism, and endometriosis. Expanded repositioning of oncology agents in order to benefit benign but serious medical illnesses is warranted.
Collapse
Affiliation(s)
- Mina Nikanjam
- Division of Hematology-Oncology, University of California, San Diego, La Jolla, CA, USA.
| | - Kaitlyn Wells
- Department of Pharmacy, University of California, San Diego, La Jolla, CA, USA
| | - Shumei Kato
- Division of Hematology-Oncology, University of California, San Diego, La Jolla, CA, USA
| | - Jacob J Adashek
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Shanna Block
- Department of Pharmacy, University of California, San Diego, La Jolla, CA, USA
| | - Razelle Kurzrock
- Division of Hematology-Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA; WIN Consortium, Chevilly-Larue, France.
| |
Collapse
|
18
|
Kast RE. IC Regimen: Delaying Resistance to Lorlatinib in ALK Driven Cancers by Adding Repurposed Itraconazole and Cilostazol. Cells 2024; 13:1175. [PMID: 39056757 PMCID: PMC11274432 DOI: 10.3390/cells13141175] [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: 06/21/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Lorlatinib is a pharmaceutical ALK kinase inhibitor used to treat ALK driven non-small cell lung cancers. This paper analyses the intersection of past published data on the physiological consequences of two unrelated drugs from general medical practice-itraconazole and cilostazol-with the pathophysiology of ALK positive non-small cell lung cancer. A conclusion from that data analysis is that adding itraconazole and cilostazol may make lorlatinib more effective. Itraconazole, although marketed worldwide as a generic antifungal drug, also inhibits Hedgehog signaling, Wnt signaling, hepatic CYP3A4, and the p-gp efflux pump. Cilostazol, marketed worldwide as a generic thrombosis preventative drug, acts by inhibiting phosphodiesterase 3, and, by so doing, lowers platelets' adhesion, thereby partially depriving malignant cells of the many tumor trophic growth factors supplied by platelets. Itraconazole may enhance lorlatinib effectiveness by (i) reducing or stopping a Hedgehog-ALK amplifying feedback loop, by (ii) increasing lorlatinib's brain levels by p-gp inhibition, and by (iii) inhibiting growth drive from Wnt signaling. Cilostazol, surprisingly, carries minimal bleeding risk, lower than that of aspirin. Risk/benefit assessment of the combination of metastatic ALK positive lung cancer being a low-survival disease with the predicted safety of itraconazole-cilostazol augmentation of lorlatinib favors a trial of this drug trio in ALK positive lung cancer.
Collapse
|
19
|
Elhassan E, Girleanu C, Kelly P, Power DG, Sweeney P, Mayer N, Bambury RM. Anaplastic Lymphoma Kinase (ALK)-Rearranged Renal Cell Carcinoma: A Case Report Highlighting Diagnostic Challenges and Therapeutic Opportunities. Cureus 2024; 16:e65621. [PMID: 39205743 PMCID: PMC11350486 DOI: 10.7759/cureus.65621] [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] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
A 57-year-old male underwent an open right radical nephrectomy in 2015 for a 3-cm kidney tumor which was classified at the time as a combined tubulocystic and collecting duct carcinoma. One of six nodes was positive for metastatic carcinoma and the patient received adjuvant carboplatin/gemcitabine chemotherapy. In 2020, he developed enlarging retroperitoneal adenopathy and underwent a retroperitoneal lymph node dissection with 11 of 13 nodes in the resected specimen positive for the previously described renal carcinoma, followed by adjuvant radiotherapy. In November 2022, he again underwent surgery for further locoregional recurrence with resection of a right psoas mass lesion and right hemicolectomy. Pathology on this occasion was reclassified as anaplastic lymphoma kinase-rearranged renal cell carcinoma (ALK-RCC). Shortly afterward, a restaging CT revealed multiple liver metastases and evidence of further disease recurrence in the right renal bed. He commenced alectinib with a complete radiological response and has continued on it for 12 months at the time of writing this report. To our knowledge, there are only five prior reports of ALK-RCC treated with targeted ALK inhibitor therapy in the literature. We report this case to highlight the importance of recognizing and diagnosing this rare RCC subtype since it has significant therapeutic implications. Furthermore, to our knowledge, this patient has had the longest follow-up reported to date in the literature so far. A concerted effort by the histopathology and oncology community is needed to gather more data on the incidence and treatment outcomes of these tumors so that progress can be made in optimizing their management. It is important to consider novel and emerging entities from the most recent WHO 2022 classification, many of which are defined by molecular characteristics with associated therapeutic implications.
Collapse
Affiliation(s)
| | | | - Paul Kelly
- Radiation Oncology, Bon Secours Hospital, Cork, IRL
| | - Derek G Power
- Medical Oncology, Cork University Hospital, Cork, IRL
| | | | - Nick Mayer
- Histopathology, Cork University Hospital, Cork, IRL
| | | |
Collapse
|
20
|
Ilié M, Goffinet S, Rignol G, Lespinet-Fabre V, Lalvée S, Bordone O, Zahaf K, Bonnetaud C, Washetine K, Lassalle S, Long-Mira E, Heeke S, Hofman V, Hofman P. Shifting from Immunohistochemistry to Screen for ALK Rearrangements: Real-World Experience in a Large Single-Center Cohort of Patients with Non-Small-Cell Lung Cancer. Cancers (Basel) 2024; 16:2219. [PMID: 38927925 PMCID: PMC11201761 DOI: 10.3390/cancers16122219] [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: 05/22/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The identification of ALK fusions in advanced non-small-cell lung carcinoma (aNSCLC) is mandatory for targeted therapy. The current diagnostic approach employs an algorithm using ALK immunohistochemistry (IHC) screening, followed by confirmation through ALK FISH and/or next-generation sequencing (NGS). Challenges arise due to the infrequency of ALK fusions (3-7% of aNSCLC), the suboptimal specificity of ALK IHC and ALK FISH, and the growing molecular demands placed on small tissue samples, leading to interpretative, tissue availability, and time-related issues. This study investigates the effectiveness of RNA NGS as a reflex test for identifying ALK fusions in NSCLC, with the goal of replacing ALK IHC in the systematic screening process. The evaluation included 1246 NSCLC cases using paired techniques: ALK IHC, ALK FISH, and ALK NGS. ALK IHC identified 51 positive cases (4%), while RNA NGS detected ALK alterations in 59 cases (4.8%). Of the 59 ALK-positive cases identified via NGS, 53 (89.8%) were confirmed to be positive. This included 51 cases detected via both FISH and IHC, and 2 cases detected only via FISH, as they were completely negative according to IHC. The combined reporting time for ALK IHC and ALK FISH averaged 13 days, whereas ALK IHC and RNA NGS reports were obtained in an average of 4 days. These results emphasize the advantage of replacing systematic ALK IHC screening with RNA NGS reflex testing for a more comprehensive and accurate assessment of ALK status.
Collapse
Affiliation(s)
- Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France;
| | - Samantha Goffinet
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France;
| | - Guylène Rignol
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France;
| | - Virginie Lespinet-Fabre
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
| | - Salomé Lalvée
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
| | - Olivier Bordone
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France;
| | - Katia Zahaf
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
| | - Christelle Bonnetaud
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France;
| | - Kevin Washetine
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France;
| | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France;
| | - Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France;
| | - Simon Heeke
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France;
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France; (M.I.); (S.G.); (G.R.); (V.L.-F.); (S.L.); (K.Z.); (C.B.); (K.W.); (S.L.); (E.L.-M.); (V.H.)
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice University Hospital, FHU OncoAge, IHU RespirERA, 06000 Nice, France;
| |
Collapse
|
21
|
Zhang W, Zhang Y, Zhou L, Tan N, Bai Y, Xing S. Primary lung adenocarcinoma with breast metastasis harboring the EML4‑ALK fusion: A case report. Oncol Lett 2024; 27:276. [PMID: 38690101 PMCID: PMC11058636 DOI: 10.3892/ol.2024.14409] [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: 01/31/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Pulmonary adenocarcinoma with breast metastasis is rarely encountered in clinical practice. Therefore, precise clinical diagnosis of patients with this disease is crucial when selecting subsequent treatment modalities and for overall prognosis assessment. The present study reported on a case of lung cancer with breast metastasis harboring the EML4-ALK fusion. The patient was initially diagnosed with triple-negative breast cancer with lung metastasis, but comprehensive breast cancer treatment was ineffective. Reevaluation of the patient's condition via lung biopsy revealed primary lung adenocarcinoma. In addition, the results of genetic testing revealed the EML4-ALK fusion protein in both lung and breast tissues. After treatment with ALK inhibitors, the patient's symptoms improved rapidly. This case highlights the prolonged diagnostic journey from presentation with a breast mass to ultimately being diagnosed with lung cancer with breast metastasis, underscoring the critical need for heightened awareness among clinicians regarding the possibility of rare metastatic patterns. Timely identification of lung cancer with breast metastasis, facilitated by comprehensive genetic testing, not only refines treatment decisions but also emphasizes the importance of interdisciplinary collaboration in navigating complex clinical scenarios. Such insight contributes to the ongoing development of personalized cancer care that guides clinicians toward more effective and tailored therapeutic strategies for patients with similar diagnostic challenges.
Collapse
Affiliation(s)
- Wenwen Zhang
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yu Zhang
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Lei Zhou
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Na Tan
- Department of Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yuju Bai
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Shiyun Xing
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
22
|
Aleksakhina SN, Ivantsov AO, Imyanitov EN. Agnostic Administration of Targeted Anticancer Drugs: Looking for a Balance between Hype and Caution. Int J Mol Sci 2024; 25:4094. [PMID: 38612902 PMCID: PMC11012409 DOI: 10.3390/ijms25074094] [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: 02/18/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Many tumors have well-defined vulnerabilities, thus potentially allowing highly specific and effective treatment. There is a spectrum of actionable genetic alterations which are shared across various tumor types and, therefore, can be targeted by a given drug irrespective of tumor histology. Several agnostic drug-target matches have already been approved for clinical use, e.g., immune therapy for tumors with microsatellite instability (MSI) and/or high tumor mutation burden (TMB), NTRK1-3 and RET inhibitors for cancers carrying rearrangements in these kinases, and dabrafenib plus trametinib for BRAF V600E mutated malignancies. Multiple lines of evidence suggest that this histology-independent approach is also reasonable for tumors carrying ALK and ROS1 translocations, biallelic BRCA1/2 inactivation and/or homologous recombination deficiency (HRD), strong HER2 amplification/overexpression coupled with the absence of other MAPK pathway-activating mutations, etc. On the other hand, some well-known targets are not agnostic: for example, PD-L1 expression is predictive for the efficacy of PD-L1/PD1 inhibitors only in some but not all cancer types. Unfortunately, the individual probability of finding a druggable target in a given tumor is relatively low, even with the use of comprehensive next-generation sequencing (NGS) assays. Nevertheless, the rapidly growing utilization of NGS will significantly increase the number of patients with highly unusual or exceptionally rare tumor-target combinations. Clinical trials may provide only a framework for treatment attitudes, while the decisions for individual patients usually require case-by-case consideration of the probability of deriving benefit from agnostic versus standard therapy, drug availability, associated costs, and other circumstances. The existing format of data dissemination may not be optimal for agnostic cancer medicine, as conventional scientific journals are understandably biased towards the publication of positive findings and usually discourage the submission of case reports. Despite all the limitations and concerns, histology-independent drug-target matching is certainly feasible and, therefore, will be increasingly utilized in the future.
Collapse
Affiliation(s)
- Svetlana N. Aleksakhina
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
| | - Alexander O. Ivantsov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| |
Collapse
|
23
|
Gouda MA, Subbiah V. Tissue-Agnostic Cancer Therapy Approvals. Surg Oncol Clin N Am 2024; 33:243-264. [PMID: 38401908 DOI: 10.1016/j.soc.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2024]
Abstract
Tumor-agnostic, or histology-agnostic, cancer therapy marks a groundbreaking evolution in the realm of precision oncology. In stark contrast to conventional cancer treatments that categorize malignancies based on their tissue of origin (eg, breast, lung, renal cell, etc), tumor-agnostic therapies transcend histologic boundaries, honing in on the genetic and molecular attributes of tumors, regardless of their location. This article offers a comprehensive review of the current landscape of tissue-agnostic cancer therapies and provides clinical insights to empower surgical oncologists with a deeper understanding of these innovative therapeutic approaches.
Collapse
Affiliation(s)
- Mohamed A Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 455, Houston, TX, USA
| | - Vivek Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute, 335 24th Avenue North Suite 300, Nashville, TN 37203, USA.
| |
Collapse
|
24
|
Adashek JJ, Kato S, Sicklick JK, Lippman SM, Kurzrock R. If it's a target, it's a pan-cancer target: Tissue is not the issue. Cancer Treat Rev 2024; 125:102721. [PMID: 38522181 PMCID: PMC11093268 DOI: 10.1016/j.ctrv.2024.102721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
Cancer is traditionally diagnosed and treated on the basis of its organ of origin (e.g., lung or colon cancer). However, organ-of-origin diagnostics does not reveal the underlying oncogenic drivers. Fortunately, molecular diagnostics have advanced at a breathtaking pace, and it is increasingly apparent that cancer is a disease of the genome. Hence, we now have multiple genomic biomarker-based, tissue-agnostic Food and Drug Administration approvals for both gene- and immune-targeted therapies (larotrectinib/entrectinib, for NTRK fusions; selpercatinib, RET fusions; dabrafenib plus trametinib, BRAFV600E mutations; pembrolizumab/dostarlimab, microsatellite instability; and pembrolizumab for high tumor mutational burden; pemigatinib is also approved for FGFR1-rearranged myeloid/lymphoid neoplasms). There are emerging targets as well, including but not limited to ALK, BRCA and/or homologous repair deficiency, ERBB2 (HER2), IDH1/2, KIT, KRASG12C, NRG1, and VHL. Many tissue-agnostic approvals center on rare/ultra-rare biomarkers (often < 1 % of cancers), necessitating screening hundreds of tumors to find a single one harboring the cognate molecular alteration. Approval has generally been based on small single-arm studies (<30-100 patients) with high response rates (>30 % to > 75 %) of remarkable durability. Because of biomarker rarity, single-gene testing is not practical; next generation sequencing of hundreds of genes must be performed to obtain timely answers. Resistance to biomarker-driven therapeutics is often due to secondary mutations or co-driver gene defects; studies are now addressing the need for customized drug combinations matched to the complex molecular alteration portfolio in each tumor. Future investigation should expand tissue-agnostic therapeutics to encompass both hematologic and solid malignancies and include biomarkers beyond those that are DNA-based.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jason K Sicklick
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA; Department of Surgery, Division of Surgical Oncology, University of California San Diego, UC San Diego Health, San Diego, CA, USA; Department of Pharmacology, University of California San Diego, UC San Diego Health, San Diego, CA, USA
| | - Scott M Lippman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee Wisconsin, USA; WIN Consortium, Paris France; University of Nebraska, United States.
| |
Collapse
|
25
|
Rosebush MS, Smith MH, Cordell KG, Callahan N, Zaid W, Gagan J, Bishop JA. Reticular Myxoid Odontogenic Neoplasm with Novel STRN::ALK Fusion: Report of 2 Cases in 3-Year-Old Males. Head Neck Pathol 2024; 18:26. [PMID: 38526831 PMCID: PMC10963713 DOI: 10.1007/s12105-024-01633-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/15/2024] [Indexed: 03/27/2024]
Abstract
Odontogenic tumors represent a collection of entities ranging from hamartomas to destructive benign and malignant neoplasms. Occasionally, pathologists encounter gnathic lesions which clearly exhibit an odontogenic origin but do not fit within the confines of established diagnoses. Here, we describe two such odontogenic tumors, both affecting 3-year-old males. Each case presented as a destructive, radiolucent mandibular lesion composed of mesenchymal cells, some with unique multi-lobed nuclei, frequently arranged in a reticular pattern and supported by a myxoid stroma with focal laminations. Production of odontogenic hard tissues was also seen. Because of their unique microscopic features, both cases were investigated by next-generation sequencing and found to harbor the same STRN::ALK oncogene fusion. To our knowledge, these cases represent the first report of an odontogenic tumor with a STRN::ALK gene rearrangement. We propose the possibility that this neoplasm could be separate from other known odontogenic tumors. Both patients were treated with surgical resection and reconstruction. The prognosis of patients with this entity is currently uncertain but shall become more apparent over time as more cases are identified and followed.
Collapse
Affiliation(s)
- Molly S Rosebush
- Louisiana State University Health Sciences Center New Orleans, 1100 Florida Avenue, New Orleans, LA, 70119, USA.
| | | | - Kitrina G Cordell
- Louisiana State University Health Sciences Center New Orleans, 1100 Florida Avenue, New Orleans, LA, 70119, USA
| | | | - Waleed Zaid
- Louisiana State University Health Sciences Center New Orleans, 1100 Florida Avenue, New Orleans, LA, 70119, USA
| | - Jeffrey Gagan
- The University of Texas Southwestern Medical Center, Dallas, USA
| | - Justin A Bishop
- The University of Texas Southwestern Medical Center, Dallas, USA
| |
Collapse
|
26
|
Mousa DPV, Mavrovounis G, Argyropoulos D, Stranjalis G, Kalamatianos T. Anaplastic Lymphoma Kinase (ALK) in Posterior Cranial Fossa Tumors: A Scoping Review of Diagnostic, Prognostic, and Therapeutic Perspectives. Cancers (Basel) 2024; 16:650. [PMID: 38339401 PMCID: PMC10854950 DOI: 10.3390/cancers16030650] [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: 12/30/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Anaplastic Lymphoma Kinase (ALK) has been implicated in several human cancers. This review aims at mapping the available literature on the involvement of ALK in non-glial tumors localized in the posterior cranial fossa and at identifying diagnostic, prognostic, and therapeutic considerations. Following the PRISMA-ScR guidelines, studies were included if they investigated ALK's role in primary CNS, non-glial tumors located in the posterior cranial fossa. A total of 210 manuscripts were selected for full-text review and 16 finally met the inclusion criteria. The review included 55 cases of primary, intracranial neoplasms with ALK genetic alterations and/or protein expression, located in the posterior fossa, comprising of medulloblastoma, anaplastic large-cell lymphoma, histiocytosis, inflammatory myofibroblastic tumors, and intracranial myxoid mesenchymal tumors. ALK pathology was investigated via immunohistochemistry or genetic analysis. Several studies provided evidence for potential diagnostic and prognostic value for ALK assessment as well as therapeutic efficacy in its targeting. The available findings on ALK in posterior fossa tumors are limited. Nevertheless, previous findings suggest that ALK assessment is of diagnostic and prognostic value in medulloblastoma (WNT-activated). Interestingly, a substantial proportion of ALK-positive/altered CNS histiocytoses thus far identified have been localized in the posterior fossa. The therapeutic potential of ALK inhibition in histiocytosis warrants further investigation.
Collapse
Affiliation(s)
| | - Georgios Mavrovounis
- Department of Neurosurgery, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41334 Larissa, Greece;
- Department of Neurosurgery, Evangelismos Hospital, School of Medicine, Faculty of Health Sciences, National and Kapodistrian University of Athens, 10676 Athens, Greece;
| | - Dionysios Argyropoulos
- Department of Psychiatry, Eginition Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - George Stranjalis
- Department of Neurosurgery, Evangelismos Hospital, School of Medicine, Faculty of Health Sciences, National and Kapodistrian University of Athens, 10676 Athens, Greece;
| | - Theodosis Kalamatianos
- Department of Neurosurgery, Evangelismos Hospital, School of Medicine, Faculty of Health Sciences, National and Kapodistrian University of Athens, 10676 Athens, Greece;
| |
Collapse
|