1
|
Yin H, Xin Y, Yang J, Luo Q, Yang M, Sun J, Wang Y, Wang Q, Kalvakolanu DV, Guo B, Jiang W, Zhang L. Multifunctional nanozymes: Promising applications in clinical diagnosis and cancer treatment. Biosens Bioelectron 2025; 279:117383. [PMID: 40121930 DOI: 10.1016/j.bios.2025.117383] [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: 03/03/2024] [Revised: 02/09/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Cancer remains one of the greatest challenges in modern medicine. Traditional chemotherapy drugs often cause severe side effects, including nausea, vomiting, diarrhea, neurotoxicity, liver damage, and nephrotoxicity. In addition to these adverse effects, high recurrence and metastasis rates following treatment pose significant challenges for clinicians. There is an urgent need for novel therapeutic strategies to improve cancer treatment outcomes. In this context, nanozymes-artificial enzyme mimetics-have attracted considerable attention due to their unique advantages, including potent tumor-killing effects, enhanced biocompatibility, and reduced toxicity. Notably, nanozymes can dynamically monitor tumors through imaging and tracing. The multifunctional nanozyme (MN) is a promising research focus, integrating multiple catalytic activities, signal enhancement, sensing capabilities, and diverse modifications within a single nanozyme system. MNs can selectively target tumor regions, facilitating synergistic effects with other cancer therapies while enabling real-time imaging and tumor tracking. In this review, we first categorize MNs based on their composition and structural characteristics. We then discuss the primary mechanisms by which MNs exert their anticancer effects. Additionally, we review three types of MN biosensors and four MN-based therapeutic approaches applied in cancer treatment. Finally, we highlight the current challenges in MN research and provide an outlook on future developments in this field.
Collapse
Affiliation(s)
- Hailin Yin
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yang Xin
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Jiaying Yang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Qian Luo
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Mei Yang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Jicheng Sun
- Department of Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Yingtong Wang
- The Undergraduate Center of Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Qi Wang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Dhan V Kalvakolanu
- Greenebaum NCI Comprehensive Cancer Center, Department of Microbiology and Immunology, University of Maryland School Medicine, Baltimore, MD, USA
| | - Baofeng Guo
- Department of Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Wei Jiang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China.
| | - Ling Zhang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| |
Collapse
|
2
|
Hsieh CH, Chen YX, Tseng TY, Li A, Huang HC, Juan HF. Transcriptionally distinct malignant neuroblastoma populations show selective response to adavosertib treatment. Neurotherapeutics 2025; 22:e00575. [PMID: 40118716 DOI: 10.1016/j.neurot.2025.e00575] [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: 03/08/2025] [Indexed: 03/23/2025] Open
Abstract
Neuroblastoma is an aggressive childhood cancer that arises from the sympathetic nervous system. Despite advances in treatment, high-risk neuroblastoma remains difficult to manage due to its heterogeneous nature and frequent development of drug resistance. Drug repurposing guided by single-cell analysis presents a promising strategy for identifying new therapeutic options. Here, we aim to characterize high-risk neuroblastoma subpopulations and identify effective repurposed drugs for targeted treatment. We performed single-cell transcriptomic analysis of neuroblastoma samples, integrating bulk RNA-seq data deconvolution with clinical outcomes to define distinct malignant cell states. Using a systematic drug repurposing pipeline, we identified and validated potential therapeutic agents targeting specific high-risk neuroblastoma subpopulations. Single-cell analysis revealed 17 transcriptionally distinct neuroblastoma subpopulations. Survival analysis identified a highly aggressive subpopulation characterized by elevated UBE2C/PTTG1 expression and poor patient outcomes, distinct from a less aggressive subpopulation with favorable prognosis. Drug repurposing screening identified Adavosertib as particularly effective against the aggressive subpopulation, validated using SK-N-DZ cells as a representative model. Mechanistically, Adavosertib suppressed cell proliferation through AKT/mTOR pathway disruption, induced G2/M phase cell cycle arrest, and promoted apoptosis. Further analysis revealed UBE2C and PTTG1 as key molecular drivers of drug resistance, where their overexpression enhanced proliferation, Adavosertib resistance, and cell migration. This study establishes a single-cell-based drug repurposing strategy for high-risk neuroblastoma treatment. Our approach successfully identified Adavosertib as a promising repurposed therapeutic agent for targeting specific high-risk neuroblastoma subpopulations, providing a framework for developing more effective personalized treatment strategies.
Collapse
Affiliation(s)
- Chiao-Hui Hsieh
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yi-Xuan Chen
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Tzu-Yang Tseng
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Albert Li
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Hsueh-Fen Juan
- Department of Life Science, National Taiwan University, Taipei, Taiwan; Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan; Center for Computational and Systems Biology, National Taiwan University, Taipei, Taiwan; Center for Advanced Computing and Imaging in Biomedicine, Taipei, Taiwan.
| |
Collapse
|
3
|
Chen H, An Y, Wang C, Zhou J. Circulating tumor DNA in colorectal cancer: biology, methods and applications. Discov Oncol 2025; 16:439. [PMID: 40167831 PMCID: PMC11961841 DOI: 10.1007/s12672-025-02220-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
In the practice of colorectal cancer (CRC), traditional tumor tissue analysis is limited by intratumoral and intertumoral heterogeneity and its invasive nature. Circulating tumor DNA (ctDNA) analysis, a promising liquid biopsy approach, has been increasingly explored in clinical studies. Biologically, ctDNA is characterized by tumor-specific diversity and rapid clearance from circulation, enabling real-time, dynamic, and repeatable assessments. Technologically, PCR- and NGS-based downstream analysis methods have been developed and validated. However, variables in pre-analytical and analytical procedures underscores the need for standardized protocols. Compared with clinicopathology-based risk stratification, ctDNA-based molecular residual disease detection has demonstrated significant potential in guiding treatment decisions. Qualitative and quantitative changes in ctDNA have also shown predictive and prognostic value during neoadjuvant or adjuvant treatment, as well as in later-line treatment for metastatic CRC. Specific molecular aberrations in ctDNA can not only assist in identifying candidates for targeted therapies but also reveal resistance mechanisms. Additionally, emerging research is exploring the potential of ctDNA in early cancer detection. Overall, as a novel biomarker, ctDNA holds substantial promise in advancing clinical practice. This review focuses on the biological characteristics, pre-analytical variables, and downstream analysis methods of ctDNA and summarizes its role across various clinical scenarios in CRC.
Collapse
Affiliation(s)
- Han Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1, Shuaifuyuan, Beijing, 100730, China
| | - Yang An
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1, Shuaifuyuan, Beijing, 100730, China
| | - Chentong Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1, Shuaifuyuan, Beijing, 100730, China
| | - Jiaolin Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1, Shuaifuyuan, Beijing, 100730, China.
| |
Collapse
|
4
|
Sabit H, Attia MG, Mohamed N, Taha PS, Ahmed N, Osama S, Abdel-Ghany S. Beyond traditional biopsies: the emerging role of ctDNA and MRD on breast cancer diagnosis and treatment. Discov Oncol 2025; 16:271. [PMID: 40050490 PMCID: PMC11885725 DOI: 10.1007/s12672-025-01940-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Breast cancer management has traditionally relied on tissue biopsies and imaging, which offer limited insights into the disease. However, the discovery of circulating tumor DNA (ctDNA) and minimal residual disease (MRD) detection has revolutionized our approach to breast cancer. ctDNA, which is fragmented tumor DNA found in the bloodstream, provides a minimally invasive way to understand the tumor's genomic landscape, revealing heterogeneity and critical mutations that biopsies may miss. MRD, which indicates cancer cells that remain after treatment, can now be detected using ctDNA and other advanced methods, improving our ability to predict disease recurrence. This allows for personalized adjuvant therapies based on individual MRD levels, avoiding unnecessary treatments for patients with low MRD. This review discusses how ctDNA and MRD represent a paradigm shift towards personalized, genomically guided cancer care, which has the potential to significantly improve patient outcomes in breast cancer.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt.
| | - Manar G Attia
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Nouran Mohamed
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Pancé S Taha
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Nehal Ahmed
- Department of Agriculture Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Salma Osama
- Department of Agriculture Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| |
Collapse
|
5
|
Takahashi N, Pongor L, Agrawal SP, Shtumpf M, Gurjar A, Rajapakse VN, Shafiei A, Schultz CW, Kim S, Roame D, Carter P, Vilimas R, Nichols S, Desai P, Figg WD, Bagheri M, Teif VB, Thomas A. Genomic alterations and transcriptional phenotypes in circulating free DNA and matched metastatic tumor. Genome Med 2025; 17:15. [PMID: 40001151 PMCID: PMC11863907 DOI: 10.1186/s13073-025-01438-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Profiling circulating cell-free DNA (cfDNA) has become a fundamental practice in cancer medicine, but the effectiveness of cfDNA at elucidating tumor-derived molecular features has not been systematically compared to standard single-lesion tumor biopsies in prospective cohorts of patients. The use of plasma instead of tissue to guide therapy is particularly attractive for patients with small cell lung cancer (SCLC), due to the aggressive clinical course of this cancer, which makes obtaining tumor biopsies exceedingly challenging. METHODS In this study, we analyzed a prospective cohort of 49 plasma samples obtained before, during, and after treatment from 20 patients with recurrent SCLC. We conducted cfDNA low-pass whole genome sequencing (0.1X coverage), comparing it with time-point matched tumor characterized using whole-exome (130X) and transcriptome sequencing. RESULTS A direct comparison of cfDNA and tumor biopsy revealed that cfDNA not only mirrors the mutation and copy number landscape of the corresponding tumor but also identifies clinically relevant resistance mechanisms and cancer driver alterations not detected in matched tumor biopsies. Longitudinal cfDNA analysis reliably tracks tumor response, progression, and clonal evolution. Sequencing coverage of plasma DNA fragments around transcription start sites showed distinct treatment-related changes and captured the expression of key transcription factors such as NEUROD1 and REST in the corresponding SCLC tumors. This allowed for the prediction of SCLC neuroendocrine phenotypes and treatment responses. CONCLUSIONS cfDNA captures a comprehensive view of tumor heterogeneity and evolution. These findings have significant implications for the non-invasive stratification of SCLC, a disease currently treated as a single entity.
Collapse
Affiliation(s)
- Nobuyuki Takahashi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Lorinc Pongor
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | | | - Mariya Shtumpf
- School of Life Sciences, University of Essex, Colchester, UK
| | - Ankita Gurjar
- School of Life Sciences, University of Essex, Colchester, UK
| | - Vinodh N Rajapakse
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Ahmad Shafiei
- Department of Radiology and Imaging Sciences, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Christopher W Schultz
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Sehyun Kim
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Diana Roame
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Paula Carter
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Rasa Vilimas
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Samantha Nichols
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Parth Desai
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - William Douglas Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Mohammad Bagheri
- Department of Radiology and Imaging Sciences, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Vladimir B Teif
- School of Life Sciences, University of Essex, Colchester, UK.
| | - Anish Thomas
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA.
| |
Collapse
|
6
|
White MG, Zeineddine MA, Fallon EA, Zeineddine FA, Dansby J, Chowdhury S, Hornstein N, Yousef A, Yousef M, Bhutiani N, Gu Y, Kee B, Dasari A, Overman MJ, Raghav K, Kopetz S, Uppal A, Taggart M, Newhook T, Fournier K, Helmink B, Drusbosky LM, Shen JP. The Landscape of ctDNA in Appendiceal Adenocarcinoma. Clin Cancer Res 2025; 31:551-560. [PMID: 39679931 PMCID: PMC11790361 DOI: 10.1158/1078-0432.ccr-24-2474] [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: 07/30/2024] [Revised: 09/23/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE Appendiceal adenocarcinoma is a rare malignancy with distinct histopathologic subtypes and a natural history with metastasis primarily limited to the peritoneum. Little is known about the molecular pathogenesis of appendiceal adenocarcinoma relative to common tumors. EXPERIMENTAL DESIGN We analyzed molecular data for patients within the Guardant Health database with appendix cancer (n = 718). We then identified patients with appendiceal adenocarcinoma at our institution (from October 2004-September 2022) for whom ctDNA mutation profiling (liquid biopsy) was performed (n = 168) and extracted clinicopathologic and outcomes data. Of these 168 patients, 57 also had tissue-based tumor mutational profiling, allowing for evaluation of concordance between liquid and tissue assays. RESULTS The mutational landscape of ctDNA in appendiceal adenocarcinoma is distinct from tissue-based sequencing, with TP53 being the most frequently mutated (46%). Relative to other tumors, appendiceal adenocarcinoma seems less likely to shed ctDNA, with only 38% of patients with metastatic appendiceal adenocarcinoma having detectable ctDNA (OR = 0.26; P < 0.0001 relative to colorectal cancer). When detectable, the median variant allele frequency was significantly lower in appendiceal adenocarcinoma (0.4% vs. 1.3% for colorectal cancer; P ≤ 0.001). High-grade, signet ring, or colonic-type histology, metastatic spread beyond the peritoneum, and TP53 mutation were associated with detectable ctDNA. With respect to clinical translation, patients with detectable ctDNA had worse overall survival (HR = 2.32; P = 0.048). In the Guardant Health cohort, actionable mutations were found in 93 patients (13.0%). CONCLUSIONS Although metastatic appendiceal adenocarcinoma tumors are less likely to shed tumor DNA into the blood relative to colorectal cancer, ctDNA profiling in appendiceal adenocarcinoma has clinical utility.
Collapse
Affiliation(s)
- Michael G. White
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mohammad A. Zeineddine
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Eleanor A. Fallon
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Fadl A. Zeineddine
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Julia Dansby
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nicholas Hornstein
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Neal Bhutiani
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yue Gu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Bryan Kee
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kanwal Raghav
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Abhineet Uppal
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Melissa Taggart
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Timothy Newhook
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Keith Fournier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Beth Helmink
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - John Paul Shen
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
7
|
Singh N, Xia W, Need E, McManus K, Huang J, Shi S, Goel S. Tumor agnostic ultrasmall nanoprobes for fluorescence-guided surgical resection in peritoneal metastasis. Eur J Nucl Med Mol Imaging 2025; 52:1149-1165. [PMID: 39446146 DOI: 10.1007/s00259-024-06950-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE Surgical excision of metastases is the only curative treatment strategy in peritoneal carcinomatosis management, and the completeness of tumor resection determines the success of the surgery. Tumor-specific fluorescence-guided probes can improve the outcomes of cytoreductive surgery and thereby prognosis. This study aimed to develop and evaluate the feasibility of fluorescently labeled ultrasmall porous silica nanoparticles (UPSN) for image-guided resection of peritoneally disseminated tumors of different origins. METHODS Ultrasmall fluorescent nanoprobes were synthesized and characterized for their physicochemical properties and stability. Tumor-specific uptake and biodistribution profiles were evaluated in syngeneic CT26 colorectal and KPC-689 pancreatic cancer murine models. The practicability of real-time optical UPSN-guided resection was examined in the CT26 colorectal cancer model using a surgical stereomicroscope. Quantitative measurements of tumor sensitivity and specificity were performed. Histopathological examination validated in vivo findings about tumor-specific accumulation and safety of ultrasmall fluorescent probes. RESULTS As-synthesized UPSNs were successfully surface modified with Cy5 or Cy3 dyes maintaining sub-15 nm size and near neutral charge which is beneficial for optimized in vivo pharmacokinetics. UPSN-Cy5 demonstrated high tumor-specific uptake and favorable biodistribution profiles in peritoneal metastasis models of CT26 and KPC tumors. Dye-conjugated UPSN enabled resection of microscopic lesions and achieved a higher tumor-to-background ratios in comparison to FDA-approved indocyanine green (ICG) dye in both models. Microscopic evaluation showed tumor localization and off-target safety profile of the UPSN-Cy5. CONCLUSION Ultrasmall fluorescent probes were effective in surgical resection of peritoneal metastases with high sensitivity and specificity, thus emerging as promising tumor agnostic agents for image-guided cancer surgery.
Collapse
Affiliation(s)
- Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Wenxi Xia
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Esther Need
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Kylee McManus
- College of Science and Honors College (Biology), University of Utah, Salt Lake City, UT, 84112, USA
| | - Jiemin Huang
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA.
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA.
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, 84112, USA.
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
8
|
MacDonald WJ, Purcell C, Pinho-Schwermann M, Stubbs NM, Srinivasan PR, El-Deiry WS. Heterogeneity in Cancer. Cancers (Basel) 2025; 17:441. [PMID: 39941808 PMCID: PMC11816170 DOI: 10.3390/cancers17030441] [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: 12/19/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Cancer heterogeneity is a major challenge in oncology, complicating diagnosis, prognostication, and treatment. The clinical heterogeneity of cancer, which leads to differential treatment outcomes between patients with histopathologically similar cancers, is attributable to molecular diversity manifesting through genetic, epigenetic, transcriptomic, microenvironmental, and host biology differences. Heterogeneity is observed between patients, individual metastases, and within individual lesions. This review discusses clinical implications of heterogeneity, emphasizing need for personalized approaches to overcome challenges posed by cancer's diverse presentations. Understanding of emerging molecular diagnostic and analytical techniques can provide a view into the multidimensional complexity of cancer heterogeneity. With over 90% of cancer-related deaths associated with metastasis, we additionally explore the role heterogeneity plays in treatment resistance and recurrence of metastatic lesions. Molecular insights from next-generation sequencing, single-cell transcriptomics, liquid biopsy technology, and artificial intelligence will facilitate the development of combination therapy regimens that can potentially induce lasting and even curative treatment outcomes.
Collapse
Affiliation(s)
- William J. MacDonald
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Connor Purcell
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Maximilian Pinho-Schwermann
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Nolan M. Stubbs
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Praveen R. Srinivasan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- The Joint Program in Cancer Biology, Brown University and Brown University Health, Providence, RI 02903, USA
- Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital, Brown University, Providence, RI 02903, USA
| |
Collapse
|
9
|
Crisafulli G. Liquid Biopsy and Challenge of Assay Heterogeneity for Minimal Residual Disease Assessment in Colon Cancer Treatment. Genes (Basel) 2025; 16:71. [PMID: 39858618 PMCID: PMC11765229 DOI: 10.3390/genes16010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
This review provides a comprehensive overview of the evolving role of minimal residual disease (MRD) for patients with Colon Cancer (CC). Currently, the standard of care for patients with non-metastatic CC is adjuvant chemotherapy (ACT) for all patients with stage III and high-risk stage II CC following surgical intervention. Despite a 5-20% improvement in long-term survival outcomes, this approach also results in a significant proportion of patients receiving ACT without any therapeutic benefit and being unnecessarily exposed to the risks of secondary side effects. This underscores an unmet clinical need for more precise stratification to distinguish patients who necessitate ACT from those who can be treated with surgery alone. By employing liquid biopsy, it is possible to discern MRD enabling the categorization of patients as MRD-positive or MRD-negative, potentially revolutionizing the management of ACT. This review aimed to examine the heterogeneity of methodologies currently available for MRD detection, encompassing the state-of-the-art technologies, their respective advantages, limitations, and the technological challenges and multi-omic approaches that can be utilized to enhance assay performance. Furthermore, a discussion was held regarding the clinical trials that employ an MRD assay focusing on the heterogeneity of the assays used. These differences in methodology, target selection, and performance risk producing inconsistent results that may not solely reflect biological/clinical differences but may be the consequence of the preferential use of particular products in studies conducted in different countries. Standardization and harmonization of MRD assays will be crucial to ensure the liquid revolution delivers reliable and clinically actionable outcomes for patients.
Collapse
|
10
|
Pietsch FL, Haag F, Ayx I, Grawe F, Vellala AK, Schoenberg SO, Froelich MF, Tharmaseelan H. Textural heterogeneity of liver lesions in CT imaging - comparison of colorectal and pancreatic metastases. Abdom Radiol (NY) 2024; 49:4295-4306. [PMID: 39115682 PMCID: PMC11522118 DOI: 10.1007/s00261-024-04511-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE Tumoral heterogeneity poses a challenge for personalized cancer treatments. Especially in metastasized cancer, it remains a major limitation for successful targeted therapy, often leading to drug resistance due to tumoral escape mechanisms. This work explores a non-invasive radiomics-based approach to capture textural heterogeneity in liver lesions and compare it between colorectal cancer (CRC) and pancreatic cancer (PDAC). MATERIALS AND METHODS In this retrospective single-center study 73 subjects (42 CRC, 31 PDAC) with 1291 liver metastases (430 CRC, 861 PDAC) were segmented fully automated on contrast-enhanced CT images by a UNet for medical images. Radiomics features were extracted using the Python package Pyradiomics. The mean coefficient of variation (CV) was calculated patient-wise for each feature to quantify the heterogeneity. An unpaired t-test identified features with significant differences in feature variability between CRC and PDAC metastases. RESULTS In both colorectal and pancreatic liver metastases, interlesional heterogeneity in imaging can be observed using quantitative imaging features. 75 second-order features were extracted to compare the varying textural characteristics. In total, 18 radiomics features showed a significant difference (p < 0.05) in their expression between the two malignancies. Out of these, 16 features showed higher levels of variability within the cohort of pancreatic metastases, which, as illustrated in a radar plot, suggests greater textural heterogeneity for this entity. CONCLUSIONS Radiomics has the potential to identify the interlesional heterogeneity of CT texture among individual liver metastases. In this proof-of-concept study for the quantification and comparison of imaging-related heterogeneity in liver metastases a variation in the extent of heterogeneity levels in CRC and PDAC liver metastases was shown.
Collapse
Affiliation(s)
- Friedrich L Pietsch
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Florian Haag
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Isabelle Ayx
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Freba Grawe
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Abhinay K Vellala
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Stefan O Schoenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Matthias F Froelich
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Hishan Tharmaseelan
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
11
|
Coelho MA, Strauss ME, Watterson A, Cooper S, Bhosle S, Illuzzi G, Karakoc E, Dinçer C, Vieira SF, Sharma M, Moullet M, Conticelli D, Koeppel J, McCarten K, Cattaneo CM, Veninga V, Picco G, Parts L, Forment JV, Voest EE, Marioni JC, Bassett A, Garnett MJ. Base editing screens define the genetic landscape of cancer drug resistance mechanisms. Nat Genet 2024; 56:2479-2492. [PMID: 39424923 PMCID: PMC11549056 DOI: 10.1038/s41588-024-01948-8] [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: 12/01/2023] [Accepted: 09/13/2024] [Indexed: 10/21/2024]
Abstract
Drug resistance is a principal limitation to the long-term efficacy of cancer therapies. Cancer genome sequencing can retrospectively delineate the genetic basis of drug resistance, but this requires large numbers of post-treatment samples to nominate causal variants. Here we prospectively identify genetic mechanisms of resistance to ten oncology drugs from CRISPR base editing mutagenesis screens in four cancer cell lines using a guide RNA library predicted to install 32,476 variants in 11 cancer genes. We identify four functional classes of protein variants modulating drug sensitivity and use single-cell transcriptomics to reveal how these variants operate through distinct mechanisms, including eliciting a drug-addicted cell state. We identify variants that can be targeted with alternative inhibitors to overcome resistance and functionally validate an epidermal growth factor receptor (EGFR) variant that sensitizes lung cancer cells to EGFR inhibitors. Our variant-to-function map has implications for patient stratification, therapy combinations and drug scheduling in cancer treatment.
Collapse
Affiliation(s)
- Matthew A Coelho
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK.
- Cancer Genome Editing, Wellcome Sanger Institute, Hinxton, UK.
- Open Targets, Cambridge, UK.
| | - Magdalena E Strauss
- EMBL-European Bioinformatics Institute, Cambridge, UK
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
- Gene Editing and Cellular Research and Development, Wellcome Sanger Institute, Hinxton, UK
- Department of Mathematics and Statistics, University of Exeter, Exeter, UK
| | - Alex Watterson
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Sarah Cooper
- Gene Editing and Cellular Research and Development, Wellcome Sanger Institute, Hinxton, UK
| | - Shriram Bhosle
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | | | - Emre Karakoc
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
- Open Targets, Cambridge, UK
| | - Cansu Dinçer
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Sara F Vieira
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
- Open Targets, Cambridge, UK
| | - Mamta Sharma
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Marie Moullet
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Daniela Conticelli
- Department of Oncology, University of Turin, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Jonas Koeppel
- Generative and Synthetic Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Katrina McCarten
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Chiara M Cattaneo
- Department of Immunology and Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vivien Veninga
- Department of Immunology and Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Gabriele Picco
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
- Open Targets, Cambridge, UK
| | - Leopold Parts
- Generative and Synthetic Genomics, Wellcome Sanger Institute, Hinxton, UK
| | | | - Emile E Voest
- Department of Immunology and Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - John C Marioni
- EMBL-European Bioinformatics Institute, Cambridge, UK
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
- Genentech, South San Francisco, CA, USA
| | - Andrew Bassett
- Gene Editing and Cellular Research and Development, Wellcome Sanger Institute, Hinxton, UK
| | - Mathew J Garnett
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK.
- Open Targets, Cambridge, UK.
| |
Collapse
|
12
|
Russolillo N, Zingaretti CC, Langella S, Fontana AP, Lo Tesoriere R, Ferrero A. GAME-SCORE predicts pathological and radiological response to chemotherapy in patients with colorectal liver metastases. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108529. [PMID: 39216236 DOI: 10.1016/j.ejso.2024.108529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Genetic And Morphological Evaluation (GAME) score is the newest prognostic model for patient with colorectal liver metastases (CRLMs). Pathological and radiological responses to neoadjuvant chemotherapy (NAC) are key factors for prognostic stratification of these patients. The present study aims to evaluate the GAME-score's ability to predict pathological and radiologic responses to NAC. METHODS CRLM patients who underwent liver resection after NAC from January 2010 to December 2021 were categorized by GAME scores: low risk (LR, 0-1), moderate risk (MR, 2-3), and high risk (HR, ≥4). Correlations between groups and radiological/pathological features were analyzed. Poor pathological response was defined as Tumor Regression Grade 4-5. RESULTS Of 1054 liver resections for CRLMs, 448 were included. GAME scores were LR: 80 (18 %), MR: 228 (51 %), and HR: 140 (31 %). In this cohort, HR-GAME scores were associated with lower pathological response (LR: 67.1 %, MR: 74.9 %, HR: 82.6 %; p = 0.010). Radiologic progression occurred in 10 % of HR patients, significantly more than in LR (3.8 %) and MR (3.5 %) groups (p = 0.011). Multivariable analysis for independent predictors of pathological response confirmed HR-GAME (RR 1.843, p=0.025) along with age higher than 70 years (RR 2.111, p=0.022) and irinotecan-based NAC (RR 3.066, p < 0.001). For radiological progression disease after NAC, the HR-GAME score (RR 2.77, p=0.016) was the only independent predictor. HR-GAME scores were also associated with higher rates of mucinous differentiation (p = 0.021), satellitosis (p = 0.001), vascular invasion (p = 0.011), and perineural invasion (p = 0.010). CONCLUSIONS GAME score category should be considered into planning of therapeutic strategy of patients with CRLMs.
Collapse
Affiliation(s)
- Nadia Russolillo
- Department of Oncological and General Surgery, Mauriziano Umberto I Hospital, Largo Turati 62, 10128, Turin, Italy.
| | - Caterina C Zingaretti
- Department of Oncological and General Surgery, Mauriziano Umberto I Hospital, Largo Turati 62, 10128, Turin, Italy
| | - Serena Langella
- Department of Oncological and General Surgery, Mauriziano Umberto I Hospital, Largo Turati 62, 10128, Turin, Italy
| | - Andrea P Fontana
- Department of Oncological and General Surgery, Mauriziano Umberto I Hospital, Largo Turati 62, 10128, Turin, Italy
| | - Roberto Lo Tesoriere
- Department of Oncological and General Surgery, Mauriziano Umberto I Hospital, Largo Turati 62, 10128, Turin, Italy
| | - Alessandro Ferrero
- Department of Oncological and General Surgery, Mauriziano Umberto I Hospital, Largo Turati 62, 10128, Turin, Italy
| |
Collapse
|
13
|
Huang FF, Di XF, Bai MH. Analysis of urine cell-free DNA in bladder cancer diagnosis by emerging bioactive technologies and materials. Front Bioeng Biotechnol 2024; 12:1458362. [PMID: 39295845 PMCID: PMC11408225 DOI: 10.3389/fbioe.2024.1458362] [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: 07/02/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024] Open
Abstract
Urinary cell-free DNA (UcfDNA) is gaining recognition as an important biomarker for diagnosing bladder cancer. UcfDNA contains tumor derived DNA sequences, making it a viable candidate for non-invasive early detection, diagnosis, and surveillance of bladder cancer. The quantification and qualification of UcfDNA have demonstrated high sensitivity and specificity in the molecular characterization of bladder cancer. However, precise analysis of UcfDNA for clinical bladder cancer diagnosis remains challenging. This review summarizes the history of UcfDNA discovery, its biological properties, and the quantitative and qualitative evaluations of UcfDNA for its clinical significance and utility in bladder cancer patients, emphasizing the critical role of UcfDNA in bladder cancer diagnosis. Emerging bioactive technologies and materials currently offer promising tools for multiple UcfDNA analysis, aiming to achieve more precise and efficient capture of UcfDNA, thereby significantly enhancing diagnostic accuracy. This review also highlights breakthroughs in detection technologies and substrates with the potential to revolutionize bladder cancer diagnosis in clinic.
Collapse
Affiliation(s)
- Fei-Fei Huang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiao-Fei Di
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Mo-Han Bai
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Leto SM, Grassi E, Avolio M, Vurchio V, Cottino F, Ferri M, Zanella ER, Borgato S, Corti G, di Blasio L, Somale D, Vara-Messler M, Galimi F, Sassi F, Lupo B, Catalano I, Pinnelli M, Viviani M, Sperti L, Mellano A, Ferrero A, Zingaretti CC, Puliafito A, Primo L, Bertotti A, Trusolino L. XENTURION is a population-level multidimensional resource of xenografts and tumoroids from metastatic colorectal cancer patients. Nat Commun 2024; 15:7495. [PMID: 39209908 PMCID: PMC11362617 DOI: 10.1038/s41467-024-51909-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
The breadth and depth at which cancer models are interrogated contribute to the successful clinical translation of drug discovery efforts. In colorectal cancer (CRC), model availability is limited by a dearth of large-scale collections of patient-derived xenografts (PDXs) and paired tumoroids from metastatic disease, where experimental therapies are typically tested. Here we introduce XENTURION, an open-science resource offering a platform of 128 PDX models from patients with metastatic CRC, along with matched PDX-derived tumoroids. Multidimensional omics analyses indicate that tumoroids retain extensive molecular fidelity with parental PDXs. A tumoroid-based trial with the anti-EGFR antibody cetuximab reveals variable sensitivities that are consistent with clinical response biomarkers, mirror tumor growth changes in matched PDXs, and recapitulate EGFR genetic deletion outcomes. Inhibition of adaptive signals upregulated by EGFR blockade increases the magnitude of cetuximab response. These findings illustrate the potential of large living biobanks, providing avenues for molecularly informed preclinical research in oncology.
Collapse
Affiliation(s)
| | - Elena Grassi
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Marco Avolio
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Valentina Vurchio
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | | | - Martina Ferri
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | | | - Sofia Borgato
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Giorgio Corti
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Laura di Blasio
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Desiana Somale
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Aptuit, an Evotec Company, Verona, Italy
| | - Marianela Vara-Messler
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
- Sanofi Belgium, Zwijnaarde, Belgium
| | - Francesco Galimi
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Francesco Sassi
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
| | - Barbara Lupo
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Irene Catalano
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
| | - Marika Pinnelli
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Marco Viviani
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Luca Sperti
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Alfredo Mellano
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
| | | | | | - Alberto Puliafito
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Luca Primo
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Andrea Bertotti
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy.
- Department of Oncology, University of Torino, Candiolo, Torino, Italy.
| | - Livio Trusolino
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy.
- Department of Oncology, University of Torino, Candiolo, Torino, Italy.
| |
Collapse
|
15
|
Tuo Z, Zhang Y, Li D, Wang Y, Wu R, Wang J, Yu Q, Ye L, Shao F, Wusiman D, Yang Y, Yoo KH, Ke M, Okoli UA, Cho WC, Heavey S, Wei W, Feng D. Relationship between clonal evolution and drug resistance in bladder cancer: A genomic research review. Pharmacol Res 2024; 206:107302. [PMID: 39004242 DOI: 10.1016/j.phrs.2024.107302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/28/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
Bladder cancer stands as a prevalent global malignancy, exhibiting notable sex-based variations in both incidence and prognosis. Despite substantial strides in therapeutic approaches, the formidable challenge of drug resistance persists. The genomic landscape of bladder cancer, characterized by intricate clonal heterogeneity, emerges as a pivotal determinant in fostering this resistance. Clonal evolution, encapsulating the dynamic transformations within subpopulations of tumor cells over time, is implicated in the emergence of drug-resistant traits. Within this review, we illuminate contemporary insights into the role of clonal evolution in bladder cancer, elucidating its influence as a driver in tumor initiation, disease progression, and the formidable obstacle of therapy resistance.
Collapse
Affiliation(s)
- Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Ying Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yetong Wang
- The Fourth Corps of Students of the Basic Medical College, Army Medical University, Chongqing 400038, China
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo City, Zhejiang Province 315211, China
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Fanglin Shao
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Yubo Yang
- Department of Urology, Three Gorges Hospital, Chongqing University, Chongqing, Wanzhou 404000, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, South Korea
| | - Mang Ke
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Uzoamaka Adaobi Okoli
- Division of Surgery & Interventional Science, University College London, London W1W 7TS, UK; Basic and Translational Cancer Research Group, Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR China.
| | - Susan Heavey
- Division of Surgery & Interventional Science, University College London, London W1W 7TS, UK.
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China; Division of Surgery & Interventional Science, University College London, London W1W 7TS, UK.
| |
Collapse
|
16
|
Blanchard CE, Gomeiz AT, Avery K, Gazzah EE, Alsubaie AM, Sikaroodi M, Chiari Y, Ward C, Sanchez J, Espina V, Petricoin E, Baldelli E, Pierobon M. Signaling dynamics in coexisting monoclonal cell subpopulations unveil mechanisms of resistance to anti-cancer compounds. Cell Commun Signal 2024; 22:377. [PMID: 39061010 PMCID: PMC11282632 DOI: 10.1186/s12964-024-01742-3] [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: 03/29/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Tumor heterogeneity is a main contributor of resistance to anti-cancer targeted agents though it has proven difficult to study. Unfortunately, model systems to functionally characterize and mechanistically study dynamic responses to treatment across coexisting subpopulations of cancer cells remain a missing need in oncology. METHODS Using single cell cloning and expansion techniques, we established monoclonal cell subpopulations (MCPs) from a commercially available epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer cell line. We then used this model sensitivity to the EGFR inhibitor osimertinib across coexisting cell populations within the same tumor. Pathway-centered signaling dynamics associated with response to treatment and morphological characteristics of the MCPs were assessed using Reverse Phase Protein Microarray. Signaling nodes differentially activated in MCPs less sensitive to treatment were then pharmacologically inhibited to identify target signaling proteins putatively implicated in promoting drug resistance. RESULTS MCPs demonstrated highly heterogeneous sensitivities to osimertinib. Cell viability after treatment increased > 20% compared to the parental line in selected MCPs, whereas viability decreased by 75% in other MCPs. Reduced treatment response was detected in MCPs with higher proliferation rates, EGFR L858R expression, activation of EGFR binding partners and downstream signaling molecules, and expression of epithelial-to-mesenchymal transition markers. Levels of activation of EGFR binding partners and MCPs' proliferation rates were also associated with response to c-MET and IGFR inhibitors. CONCLUSIONS MCPs represent a suitable model system to characterize heterogeneous biomolecular behaviors in preclinical studies and identify and functionally test biological mechanisms associated with resistance to targeted therapeutics.
Collapse
Affiliation(s)
- Claire E Blanchard
- School of Systems Biology, George Mason University, 10920 George Mason Circle, Room 2016, Manassas, VA, 20110, USA
| | - Alison T Gomeiz
- School of Systems Biology, George Mason University, 10920 George Mason Circle, Room 2016, Manassas, VA, 20110, USA
| | - Kyle Avery
- School of Systems Biology, George Mason University, 10920 George Mason Circle, Room 2016, Manassas, VA, 20110, USA
| | - Emna El Gazzah
- School of Systems Biology, George Mason University, 10920 George Mason Circle, Room 2016, Manassas, VA, 20110, USA
| | - Abduljalil M Alsubaie
- School of Systems Biology, George Mason University, 10920 George Mason Circle, Room 2016, Manassas, VA, 20110, USA
| | - Masoumeh Sikaroodi
- Microbiome Analysis Center, George Mason University, Manassas, VA, 20110, USA
| | - Ylenia Chiari
- Department of Biology, George Mason University, Fairfax, VA, 22030, USA
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2TQ, UK
| | - Chelsea Ward
- School of Systems Biology, George Mason University, 10920 George Mason Circle, Room 2016, Manassas, VA, 20110, USA
| | - Jonathan Sanchez
- School of Systems Biology, George Mason University, 10920 George Mason Circle, Room 2016, Manassas, VA, 20110, USA
| | - Virginia Espina
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA
| | - Emanuel Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA
| | - Elisa Baldelli
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA
| | - Mariaelena Pierobon
- School of Systems Biology, George Mason University, 10920 George Mason Circle, Room 2016, Manassas, VA, 20110, USA.
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA.
| |
Collapse
|
17
|
Wang K, Jiang M, Li T, Liu Y, Zong Q, Xu Q, Ullah I, Chen Y, Xue W, Yuan Y. A Synergistic Chemoimmunotherapy System Leveraging PD-L1 Blocking and Bioorthogonal Prodrug Activation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402322. [PMID: 38718226 DOI: 10.1002/adma.202402322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/05/2024] [Indexed: 05/15/2024]
Abstract
Novel strategies to facilitate tumor-specific drug delivery and restore immune attacks remain challenging in overcoming the current limitations of chemoimmunotherapy. An antitumor chemoimmunotherapy system comprising bioorthogonal reaction-ready group tetrazine (TZ) modified with an anti-PD-L1 antibody (αPD-L1TZ) and TZ-activatable prodrug vinyl ether-doxorubicin (DOX-VE) for self-reinforced anti-tumor chemoimmunotherapy is proposed. The αPD-L1TZ effectively disrupts the PD-L1/PD-1 interaction and activates the DOX prodrug in situ through the bioorthogonal click reaction of TZ and VE. Conversely, the activated DOX upregulates PD-L1 on the surface of tumor cells, facilitating tumor accumulation of αPD-L1TZ and enhancing DOX-VE activation. Furthermore, the activated DOX-induced immunogenic cell death of tumor cells, substantially improving the response efficiency of αPD-L1 in an immune-suppressive tumor microenvironment. Thus, PD-L1 blocking and bioorthogonal in situ prodrug activation synergistically enhance the antitumor efficacy of the chemoimmunotherapy system. Therefore, the system significantly enhances αPD-L1 tumor accumulation and prodrug activation and induces a robust immunological memory effect to prevent tumor recurrence and metastasis. Thus, a feasible chemoimmunotherapy combination regimen is presented.
Collapse
Affiliation(s)
- Kewei Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, P. R. China
| | - Maolin Jiang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Tao Li
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
| | - Ye Liu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
| | - Qingyu Zong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
| | - Qing Xu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
| | - Ihsan Ullah
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
| | - Yahui Chen
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, P. R. China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, P. R. China
| | - Youyong Yuan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
18
|
Takahashi N, Pongor L, Agrawal SP, Shtumpf M, Rajapakse VN, Shafiei A, Schultz CW, Kim S, Roame D, Carter P, Vilimas R, Nichols S, Desai P, Figg WD, Bagheri M, Teif VB, Thomas A. Genomic alterations and transcriptional phenotypes in circulating tumor DNA and matched metastatic tumor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.597054. [PMID: 38895436 PMCID: PMC11185519 DOI: 10.1101/2024.06.02.597054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Background Profiling circulating cell-free DNA (cfDNA) has become a fundamental practice in cancer medicine, but the effectiveness of cfDNA at elucidating tumor-derived molecular features has not been systematically compared to standard single-lesion tumor biopsies in prospective cohorts of patients. The use of plasma instead of tissue to guide therapy is particularly attractive for patients with small cell lung cancer (SCLC), a cancer whose aggressive clinical course making it exceedingly challenging to obtain tumor biopsies. Methods Here, a prospective cohort of 49 plasma samples obtained before, during, and after treatment from 20 patients with recurrent SCLC, we study cfDNA low pass whole genome (0.1X coverage) and exome (130X) sequencing in comparison with time-point matched tumor, characterized using exome and transcriptome sequencing. Results Direct comparison of cfDNA versus tumor biopsy reveals that cfDNA not only mirrors the mutation and copy number landscape of the corresponding tumor but also identifies clinically relevant resistance mechanisms and cancer driver alterations not found in matched tumor biopsies. Longitudinal cfDNA analysis reliably tracks tumor response, progression, and clonal evolution. Genomic sequencing coverage of plasma DNA fragments around transcription start sites shows distinct treatment-related changes and captures the expression of key transcription factors such as NEUROD1 and REST in the corresponding SCLC tumors, allowing prediction of SCLC neuroendocrine phenotypes and treatment responses. Conclusions These findings have important implications for non-invasive stratification and subtype-specific therapies for patients with SCLC, now treated as a single disease.
Collapse
Affiliation(s)
- Nobuyuki Takahashi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
- Medical Oncology Branch, Center Hospital, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Medical Oncology, National Cancer Center East Hospital, Kashiwa, Japan
| | - Lorinc Pongor
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | | | - Mariya Shtumpf
- School of Life Sciences, University of Essex, Colchester, UK
| | - Vinodh N Rajapakse
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Ahmad Shafiei
- Department of Radiology and Imaging Sciences, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Christopher W Schultz
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Sehyun Kim
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Diana Roame
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Paula Carter
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Rasa Vilimas
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Samantha Nichols
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Parth Desai
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - William Douglas Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Mohammad Bagheri
- Department of Radiology and Imaging Sciences, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Vladimir B Teif
- School of Life Sciences, University of Essex, Colchester, UK
| | - Anish Thomas
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| |
Collapse
|
19
|
Devalle S, Aran V, Bastos Júnior CDS, Pannain VL, Brackmann P, Gregório ML, Ferreira Manso JE, Moura Neto V. A panorama of colon cancer in the era of liquid biopsy. THE JOURNAL OF LIQUID BIOPSY 2024; 4:100148. [PMID: 40027146 PMCID: PMC11863817 DOI: 10.1016/j.jlb.2024.100148] [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: 02/19/2024] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 03/05/2025]
Abstract
Colon cancer (CC) is one of the most frequent cancers worldwide being responsible for over 500 thousand deaths in 2022. Its financial and human burden is expected to increase in the next decades accompanying the growing and aging of the global population. Much of this burden could be alleviated considering that the lethality of CC is mostly due to its late diagnosis and failure in the individualized management of patients. Coordinated government actions and implementation of better diagnostic tools capable of detecting CC earlier and of tracking tumoral evolution are mandatory to achieve a reduction in CC's social impact. CtDNA-based liquid biopsy (LB) has great potential to contribute to patients' screening adhesion, CC earlier detection, and to longitudinal tumor follow-up. In this review, we will discuss the latest epidemiological data on CC disease, diagnostic, subtypes, genetics, and treatment management focusing on the advantages and limitations of ctDNA-based LB, including important bottlenecks and solutions necessary for its clinical translation. The latest ctDNA-directed CC clinical trials will also be examined.
Collapse
Affiliation(s)
- Sylvie Devalle
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Veronica Aran
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | - Vera Lucia Pannain
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo Brackmann
- Clínica de Coloproctologia do Hospital Naval Marcílio Dias - IPB/HNMD, Rio de Janeiro, Brazil
| | - Marcelo Leal Gregório
- Instituto de Pesquisas Biomédicas do Hospital Naval Marcílio Dias - IPB/HNMD, Rio de Janeiro, Brazil
| | - José Eduardo Ferreira Manso
- Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivaldo Moura Neto
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Park BC, Soh JO, Choi HJ, Park HS, Lee SM, Fu HE, Kim MS, Ko MJ, Koo TM, Lee JY, Kim YK, Lee JH. Ultrasensitive and Rapid Circulating Tumor DNA Liquid Biopsy Using Surface-Confined Gene Amplification on Dispersible Magnetic Nano-Electrodes. ACS NANO 2024; 18:12781-12794. [PMID: 38733343 DOI: 10.1021/acsnano.3c12266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2024]
Abstract
Circulating tumor DNA (ctDNA) detection has been acknowledged as a promising liquid biopsy approach for cancer diagnosis, with various ctDNA assays used for early detection and treatment monitoring. Dispersible magnetic nanoparticle-based electrochemical detection methods have been proposed as promising candidates for ctDNA detection based on the detection performance and features of the platform material. This study proposes a nanoparticle surface-localized genetic amplification approach by integrating Fe3O4-Au core-shell nanoparticles into polymerase chain reactions (PCR). These highly dispersible and magnetically responsive superparamagnetic nanoparticles act as nano-electrodes that amplify and accumulate target ctDNA in situ on the nanoparticle surface upon PCR amplification. These nanoparticles are subsequently captured and subjected to repetitive electrochemical measurements to induce reconfiguration-mediated signal amplification for ultrasensitive (∼3 aM) and rapid (∼7 min) metastatic breast cancer ctDNA detection in vitro. The detection platform can also detect metastatic biomarkers from in vivo samples, highlighting the potential for clinical applications and further expansion to rapid and ultrasensitive multiplex detection of various cancers.
Collapse
Affiliation(s)
- Bum Chul Park
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109-2136, United States
| | - Jeong Ook Soh
- Department of Bionano Engineering, Hanyang University, Ansan 15588, Republic of Korea
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Hee-Joo Choi
- Department of Pathology, Hanyang University, Seoul 04763, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul 04763, Republic of Korea
- Hanyang Biomedical Research Institute (HBRI), Hanyang University, Seoul 04763, Republic of Korea
| | - Hyeon Su Park
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Sang Min Lee
- Department of Bionano Engineering, Hanyang University, Ansan 15588, Republic of Korea
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Hong En Fu
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Myeong Soo Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Min Jun Ko
- Department of Radiology, Northwestern University, Chicago, Illinois 60611, United States
| | - Thomas Myeongseok Koo
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Jeong-Yeon Lee
- Department of Pathology, Hanyang University, Seoul 04763, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul 04763, Republic of Korea
| | - Young Keun Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
- Brain Korea Center for Smart Materials and Devices, Korea University, Seoul 02841, Republic of Korea
| | - Ju Hun Lee
- Department of Bionano Engineering, Hanyang University, Ansan 15588, Republic of Korea
- Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| |
Collapse
|
21
|
Si Y, Wang X, Su X, Weng Z, Hu Q, Li Q, Fan C, Zhang DY, Wang Y, Luo S, Song P. Extended Enrichment for Ultrasensitive Detection of Low-Frequency Mutations by Long Blocker Displacement Amplification. Angew Chem Int Ed Engl 2024; 63:e202400551. [PMID: 38416545 DOI: 10.1002/anie.202400551] [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: 01/15/2024] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 02/29/2024]
Abstract
Detecting low-frequency DNA mutations hotspots cluster is critical for cancer diagnosis but remains challenging. Quantitative PCR (qPCR) is constrained by sensitivity, and allele-specific PCR is restricted by throughput. Here we develop a long blocker displacement amplification (LBDA) coupled with qPCR for ultrasensitive and multiplexed variants detection. By designing long blocker oligos to perfectly match wildtype sequences while mispairing with mutants, long blockers enable 14-44 nt enrichment regions which is 2-fold longer than normal BDA in the experiments. For wild template with a specific nucleotide, LBDA can detect different mutation types down to 0.5 % variant allele frequency (VAF) in one reaction, with median enrichment fold of 1,000 on 21 mutant DNA templates compared to the wild type. We applied LBDA-qPCR to detect KRAS and NRAS mutation hotspots, utilizing a single plex assay capable of covering 81 mutations and tested in synthetic templates and colorectal cancer tissue samples. Moreover, the mutation types were verified through Sanger sequencing, demonstrating concordance with results obtained from next generation sequencing. Overall, LBDA-qPCR provides a simple yet ultrasensitive approach for multiplexed detection of low VAF mutations hotspots, presenting a powerful tool for cancer diagnosis and monitoring.
Collapse
Affiliation(s)
- Yunpei Si
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiawen Wang
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinglei Su
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- Institute of Molecular Medicine (IMM) Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhi Weng
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qiongzheng Hu
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250014, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | | | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Shihua Luo
- Department of Traumatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Ping Song
- School of Biomedical Engineering, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
22
|
Zarkavelis G, Amylidi AL, Torounidou N, Yerolatsite M, Keravasili A, Keramisanou V, Mauri D. Exploring RAS mutation incidence and temporal heterogeneity in metastatic colorectal cancer patients - a single-institution experience utilising circulating tumour DNA. Contemp Oncol (Pozn) 2024; 28:45-50. [PMID: 38800532 PMCID: PMC11117156 DOI: 10.5114/wo.2024.138899] [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: 02/14/2024] [Accepted: 03/25/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Colorectal cancer (CRC) remains a significant global health challenge, ranking among the leading causes of neoplastic mortality. Despite transformative therapeutic advances, a considerable proportion of patients are diagnosed with metastatic disease, and 15-30% of those initially presenting with early-stage CRC eventually experience recurrence. Comprehensive molecular testing, especially the evaluation of microsatellite instability and mutations in KRAS/NRAS or BRAF genes, is essential upon diagnosis of stage IV disease, guiding treatment decisions. Material and methods This manuscript explores the mutational landscape of KRAS and NRAS in patients with CRC, employing digital polymerase chain reaction (PCR) BEAMing for the detection of mutations in liquid biopsy. Our study enrolled patients with histologically confirmed CRC and stage IV disease, focusing on identifying mutations in KRAS and NRAS genes during various stages of therapy. Results Evaluating baseline, midline, and progression samples, we found that 66.6% maintained consistent mutational status post-disease progression, while 33.3% exhibited a shift in mutational status. The application of techniques with high sensitivity, such as BEAMing Digital PCR, is pivotal for accurate circulating tumour DNA (ctDNA) mutation detection. The study underscores the significance of continuous molecular monitoring in guiding therapeutic decisions for patients with metastatic CRC. Conclusions Our findings contribute to our understanding of the evolving mutational landscape and the potential clinical implications of ctDNA ana- lysis in the era of personalised cancer medicine.
Collapse
Affiliation(s)
- George Zarkavelis
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | - Anna Lea Amylidi
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | - Nanteznta Torounidou
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Melina Yerolatsite
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Athanasia Keravasili
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Varvara Keramisanou
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Davide Mauri
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| |
Collapse
|
23
|
Santoro-Fernandes V, Huff DT, Rivetti L, Deatsch A, Schott B, Perlman SB, Jeraj R. An automated methodology for whole-body, multimodality tracking of individual cancer lesions. Phys Med Biol 2024; 69:085012. [PMID: 38457838 DOI: 10.1088/1361-6560/ad31c6] [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: 05/24/2023] [Accepted: 03/08/2024] [Indexed: 03/10/2024]
Abstract
Objective. Manual analysis of individual cancer lesions to assess disease response is clinically impractical and requires automated lesion tracking methodologies. However, no methodology has been developed for whole-body individual lesion tracking, across an arbitrary number of scans, and acquired with various imaging modalities.Approach. This study introduces a lesion tracking methodology and benchmarked it using 2368Ga-DOTATATE PET/CT and PET/MR images of eight neuroendocrine tumor patients. The methodology consists of six steps: (1) alignment of multiple scans via image registration, (2) body-part labeling, (3) automatic lesion-wise dilation, (4) clustering of lesions based on local lesion shape metrics, (5) assignment of lesion tracks, and (6) output of a lesion graph. Registration performance was evaluated via landmark distance, lesion matching accuracy was evaluated between each image pair, and lesion tracking accuracy was evaluated via identical track ratio. Sensitivity studies were performed to evaluate the impact of lesion dilation (fixed versus automatic dilation), anatomic location, image modalities (inter- versus intra-modality), registration mode (direct versus indirect registration), and track size (number of time-points and lesions) on lesion matching and tracking performance.Main results. Manual contouring yielded 956 lesions, 1570 lesion-matching decisions, and 493 lesion tracks. The median residual registration error was 2.5 mm. The automatic lesion dilation led to 0.90 overall lesion matching accuracy, and an 88% identical track ratio. The methodology is robust regarding anatomic locations, image modalities, and registration modes. The number of scans had a moderate negative impact on the identical track ratio (94% for 2 scans, 91% for 3 scans, and 81% for 4 scans). The number of lesions substantially impacted the identical track ratio (93% for 2 nodes versus 54% for ≥5 nodes).Significance. The developed methodology resulted in high lesion-matching accuracy and enables automated lesion tracking in PET/CT and PET/MR.
Collapse
Affiliation(s)
- Victor Santoro-Fernandes
- School of Medicine and Public Health, Department of Medical Physics, University of Wisconsin, Madison, WI, United States of America
| | - Daniel T Huff
- School of Medicine and Public Health, Department of Medical Physics, University of Wisconsin, Madison, WI, United States of America
| | - Luciano Rivetti
- Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
| | - Alison Deatsch
- School of Medicine and Public Health, Department of Medical Physics, University of Wisconsin, Madison, WI, United States of America
| | - Brayden Schott
- School of Medicine and Public Health, Department of Medical Physics, University of Wisconsin, Madison, WI, United States of America
| | - Scott B Perlman
- School of Medicine and Public Health, Department of Radiology, Section of Nuclear Medicine, University of Wisconsin, Madison, WI, United States of America
| | - Robert Jeraj
- School of Medicine and Public Health, Department of Medical Physics, University of Wisconsin, Madison, WI, United States of America
- Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
24
|
Ciardiello D, Martinelli E, Troiani T, Mauri G, Rossini D, Martini G, Napolitano S, Famiglietti V, Del Tufo S, Masi G, Santini D, Avallone A, Pietrantonio F, Lonardi S, Di Maio M, Zampino MG, Fazio N, Bardelli A, Siena S, Cremolini C, Sartore-Bianchi A, Ciardiello F. Anti-EGFR Rechallenge in Patients With Refractory ctDNA RAS/BRAF wt Metastatic Colorectal Cancer: A Nonrandomized Controlled Trial. JAMA Netw Open 2024; 7:e245635. [PMID: 38592721 PMCID: PMC11004834 DOI: 10.1001/jamanetworkopen.2024.5635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/12/2024] [Indexed: 04/10/2024] Open
Abstract
Importance The available evidence regarding anti-epidermal growth factor receptor (EGFR) inhibitor rechallenge in patients with refractory circulating tumor DNA (ctDNA) RAS/BRAF wild-type (wt) metastatic colorectal cancer (mCRC) is derived from small retrospective and prospective studies. Objective To evaluate the efficacy of anti-EGFR rechallenge in patients with refractory ctDNA RAS/BRAF wt mCRC. Design, Setting, and Participants This nonrandomized controlled trial used a pooled analysis of individual patient data from patients with RAS/BRAF wt ctDNA mCRC enrolled in 4 Italian trials (CAVE, VELO, CRICKET, and CHRONOS) and treated with anti-EGFR rechallenge between 2015 and 2022 (median [IQR] follow-up, 28.1 [25.8-35.0] months). Intervention Patients received anti-EGFR rechallenge therapy, including cetuximab plus avelumab, trifluridine-tipiracil plus panitumumab, irinotecan plus cetuximab, or panitumumab monotherapy. Main Outcomes and Measures Overall survival (OS), progression-free survival (PFS), overall response rate (ORR), and disease control rate (DCR) were calculated. Exploratory subgroup analysis evaluating several clinical variables was performed. Safety was reported. Results Overall, 114 patients with RAS/BRAF wt ctDNA mCRC (median [IQR] age, 61 [29-88] years; 66 men [57.9%]) who received anti-EGFR rechallenge as experimental therapy (48 received cetuximab plus avelumab, 26 received trifluridine-tipiracil plus panitumumab, 13 received irinotecan plus cetuximab, and 27 received panitumumab monotherapy) were included in the current analysis. Eighty-three patients (72.8%) had received 2 previous lines of therapy, and 31 patients (27.2%) had received 3 or more previous lines of therapy. The ORR was 17.5% (20 patients), and the DCR was 72.3% (82 patients). The median PFS was 4.0 months (95% CI, 3.2-4.7 months), and the median OS was 13.1 months (95% CI, 9.5-16.7 months). The subgroup of patients without liver involvement had better clinical outcomes. The median PFS was 5.7 months (95% CI, 4.8-6.7 months) in patients without liver metastasis compared with 3.6 months (95% CI, 3.3-3.9 months) in patients with liver metastasis (hazard ratio, 0.56; 95% CI, 0.37-0.83; P = .004). The median OS was 17.7 months (95% CI, 13-22.4 months) in patients without liver metastasis compared with 11.5 months (95% CI, 9.3-13.9 months) in patients with liver metastasis (hazard ratio, 0.63; 95% CI, 0.41-0.97; P = .04). Treatments showed manageable toxic effects. Conclusions and Relevance These findings suggest that anti-EGFR rechallenge therapy has promising antitumor activity in patients with refractory ctDNA RAS/BRAF wt mCRC. Within the limitation of a subgroup analysis, the absence of liver metastases was associated with significant improved survival. Trial Registration ClinicalTrials.gov Identifiers: NCT02296203; NCT04561336; NCT03227926; NCT05468892.
Collapse
Affiliation(s)
- Davide Ciardiello
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Erika Martinelli
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Gianluca Mauri
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
- Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milano, Italy
- IFOM ETS–The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Daniele Rossini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Division of Medical Oncology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Giulia Martini
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Stefania Napolitano
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Vincenzo Famiglietti
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Sara Del Tufo
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Gianluca Masi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Division of Medical Oncology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Daniele Santini
- Medical Oncology Department, La Sapienza University of Rome, Rome, Italy
| | - Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–IRCCS–Fondazione G. Pascale, Napoli, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Lonardi
- Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, Molinette Hospital, Turin, Italy
| | - Maria Giulia Zampino
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Alberto Bardelli
- IFOM ETS–The AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology, Università degli Studi di Torino, Turin, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
- Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Division of Medical Oncology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
- Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
25
|
Yousafzai NA, El Khalki L, Wang W, Szpendyk J, Sossey-Alaoui K. Advances in 3D Culture Models to Study Exosomes in Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:883. [PMID: 38473244 PMCID: PMC10931050 DOI: 10.3390/cancers16050883] [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: 01/16/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Breast cancer, a leading cause of cancer-related deaths globally, exhibits distinct subtypes with varying pathological, genetic, and clinical characteristics. Despite advancements in breast cancer treatments, its histological and molecular heterogeneity pose a significant clinical challenge. Triple-negative breast cancer (TNBC), a highly aggressive subtype lacking targeted therapeutics, adds to the complexity of breast cancer treatment. Recent years have witnessed the development of advanced 3D culture technologies, such as organoids and spheroids, providing more representative models of healthy human tissue and various malignancies. These structures, resembling organs in structure and function, are generated from stem cells or organ-specific progenitor cells via self-organizing processes. Notably, 3D culture systems bridge the gap between 2D cultures and in vivo studies, offering a more accurate representation of in vivo tumors' characteristics. Exosomes, small nano-sized molecules secreted by breast cancer and stromal/cancer-associated fibroblast cells, have garnered significant attention. They play a crucial role in cell-to-cell communication, influencing tumor progression, invasion, and metastasis. The 3D culture environment enhances exosome efficiency compared to traditional 2D cultures, impacting the transfer of specific cargoes and therapeutic effects. Furthermore, 3D exosomes have shown promise in improving therapeutic outcomes, acting as potential vehicles for cancer treatment administration. Studies have demonstrated their role in pro-angiogenesis and their innate therapeutic potential in mimicking cellular therapies without side effects. The 3D exosome model holds potential for addressing challenges associated with drug resistance, offering insights into the mechanisms underlying multidrug resistance and serving as a platform for drug screening. This review seeks to emphasize the crucial role of 3D culture systems in studying breast cancer, especially in understanding the involvement of exosomes in cancer pathology.
Collapse
Affiliation(s)
- Neelum Aziz Yousafzai
- MetroHealth System, Cleveland, OH 44109, USA; (N.A.Y.); (L.E.K.); (W.W.)
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106-4909, USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106-7285, USA
| | - Lamyae El Khalki
- MetroHealth System, Cleveland, OH 44109, USA; (N.A.Y.); (L.E.K.); (W.W.)
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106-4909, USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106-7285, USA
| | - Wei Wang
- MetroHealth System, Cleveland, OH 44109, USA; (N.A.Y.); (L.E.K.); (W.W.)
- Case Comprehensive Cancer Center, Cleveland, OH 44106-7285, USA
| | - Justin Szpendyk
- MetroHealth System, Cleveland, OH 44109, USA; (N.A.Y.); (L.E.K.); (W.W.)
| | - Khalid Sossey-Alaoui
- MetroHealth System, Cleveland, OH 44109, USA; (N.A.Y.); (L.E.K.); (W.W.)
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106-4909, USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106-7285, USA
| |
Collapse
|
26
|
Walmsley CS, Jonsson P, Cheng ML, McBride S, Kaeser C, Vargas HA, Laudone V, Taylor BS, Kappagantula R, Baez P, Richards AL, Noronha AM, Perera D, Berger M, Solit DB, Iacobuzio-Donahue CA, Scher HI, Donoghue MTA, Abida W, Schram AM. Convergent evolution of BRCA2 reversion mutations under therapeutic pressure by PARP inhibition and platinum chemotherapy. NPJ Precis Oncol 2024; 8:34. [PMID: 38355834 PMCID: PMC10866935 DOI: 10.1038/s41698-024-00526-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/30/2024] [Indexed: 02/16/2024] Open
Abstract
Reversion mutations that restore wild-type function of the BRCA gene have been described as a key mechanism of resistance to Poly(ADP-ribose) polymerase (PARP) inhibitor therapy in BRCA-associated cancers. Here, we report a case of a patient with metastatic castration-resistant prostate cancer (mCRPC) with a germline BRCA2 mutation who developed acquired resistance to PARP inhibition. Extensive genomic interrogation of cell-free DNA (cfDNA) and tissue at baseline, post-progression, and postmortem revealed ten unique BRCA2 reversion mutations across ten sites. While several of the reversion mutations were private to a specific site, nine out of ten tumors contained at least one mutation, suggesting a powerful clonal selection for reversion mutations in the presence of therapeutic pressure by PARP inhibition. Variable cfDNA shed was seen across tumor sites, emphasizing a potential shortcoming of cfDNA monitoring for PARPi resistance. This report provides a genomic portrait of the temporal and spatial heterogeneity of prostate cancer under the selective pressure of a PARP inhibition and exposes limitations in the current strategies for detection of reversion mutations.
Collapse
Grants
- P30 CA008748 NCI NIH HHS
- Grant funding from ASCO Conquer Cancer Foundation CDA, NCI P30CA008748 CCITLA, Memorial Sloan Kettering Cancer Center Support Grant (P30 CA008748).
- WA has received honoraria from Roche, Medscape, Aptitude Health, Clinical Education Alliance, OncLive/MJH Life Sciences, touchIME, Pfizer, and the MedNet. WA has also received advisory board compensation from Clovis Oncology, ORIC pharmaceuticals, Daiichi Sankyo, AstraZeneca/MedImmune, Pfizer and Laekna Therapeutics, and research funding from AstraZeneca, Zenith Epigenetics, Clovis Oncology, ORIC Pharmaceuticals, Epizyme, Nuvation Bio, Merus, and Transthera.
Collapse
Affiliation(s)
- Charlotte S Walmsley
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Philip Jonsson
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Michael L Cheng
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Sean McBride
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | | | - Vincent Laudone
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | | | - Priscilla Baez
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | | | - Dilmi Perera
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Michael Berger
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - David B Solit
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | - Howard I Scher
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | - Wassim Abida
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Alison M Schram
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| |
Collapse
|
27
|
Yang Y, Wang J, Wang J, Zhao X, Zhang T, Yang Y, Pang J, Ou Q, Wu L, Xu X, Xu K, Zhao J, Bai N, Yang P, Wang S, Wang L, Bi N. Unrevealing the therapeutic benefits of radiotherapy and consolidation immunotherapy using ctDNA-defined tumor clonality in unresectable locally advanced non-small cell lung cancer. Cancer Lett 2024; 582:216569. [PMID: 38101608 DOI: 10.1016/j.canlet.2023.216569] [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: 09/07/2023] [Revised: 10/30/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023]
Abstract
Progression occurs in approximately two-thirds of patients with locally advanced non-small cell lung cancer (LA-NSCLC) receiving chemoradiation and consolidation immunotherapy. Molecular indicators for outcome prediction are under development. A novel metric, the ratio of mean to max variant allele frequency (mmVAF), was derived from 431 pre-treatment tissue biopsies from The Cancer Genome Atlas and evaluated in serial circulating tumor DNA (ctDNA) from 70 LA-NSCLC patients receiving definitive radiotherapy/chemoradiotherapy (RT/CRT) with/without immunotherapy. High mmVAFs in pre-treatment tissue biopsies, indicating clonal predominant tumors (P < 0.01), were associated with inferior overall survival [OS, hazard ratio (HR): 1.48, 95 % confidence interval (CI): 1.11-1.98]. Similar associations of mmVAF with clonality (P < 0.01) and OS (HR: 2.24, 95 % CI: 0.71-7.08) were observed in pre-treatment ctDNA. At 1-month post-RT, ctDNA mmVAF-high patients receiving consolidation immunotherapy exhibited improved progression-free survival (PFS) compared to those who did not (HR: 0.14, 95 % CI: 0.03-0.67). From the baseline to week 4 of RT and/or 1-month post-RT, survival benefits from consolidation immunotherapy were exclusively observed in ctDNA mmVAF-increased patients (PFS, HR: 0.39, 95 % CI: 0.14-1.15), especially in terms of distant metastasis (HR: 0.11, 95 % CI: 0.01-0.95). In summary, our longitudinal data demonstrated the applicability of ctDNA-defined clonality for prognostic stratification and immunotherapy benefit prediction in LA-NSCLC.
Collapse
Affiliation(s)
- Yufan Yang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jianyang Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingbo Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaotian Zhao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Tao Zhang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yin Yang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaohui Pang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Qiuxiang Ou
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Linfang Wu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Xu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kunpeng Xu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jingjing Zhao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Bai
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Peng Yang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Sha Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Luhua Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China.
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
28
|
Chuang YM, Tzeng SF, Ho PC, Tsai CH. Immunosurveillance encounters cancer metabolism. EMBO Rep 2024; 25:471-488. [PMID: 38216787 PMCID: PMC10897436 DOI: 10.1038/s44319-023-00038-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 12/02/2023] [Accepted: 12/12/2023] [Indexed: 01/14/2024] Open
Abstract
Tumor cells reprogram nutrient acquisition and metabolic pathways to meet their energetic, biosynthetic, and redox demands. Similarly, metabolic processes in immune cells support host immunity against cancer and determine differentiation and fate of leukocytes. Thus, metabolic deregulation and imbalance in immune cells within the tumor microenvironment have been reported to drive immune evasion and to compromise therapeutic outcomes. Interestingly, emerging evidence indicates that anti-tumor immunity could modulate tumor heterogeneity, aggressiveness, and metabolic reprogramming, suggesting that immunosurveillance can instruct cancer progression in multiple dimensions. This review summarizes our current understanding of how metabolic crosstalk within tumors affects immunogenicity of tumor cells and promotes cancer progression. Furthermore, we explain how defects in the metabolic cascade can contribute to developing dysfunctional immune responses against cancers and discuss the contribution of immunosurveillance to these defects as a feedback mechanism. Finally, we highlight ongoing clinical trials and new therapeutic strategies targeting cellular metabolism in cancer.
Collapse
Affiliation(s)
- Yu-Ming Chuang
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Sheue-Fen Tzeng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
| | - Chin-Hsien Tsai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.
- Department and Graduate Institute of Biochemistry, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
29
|
Wu Q, Ellis H, Siravegna G, Michel AG, Norden BL, Fece de la Cruz F, Balasooriya ER, Zhen Y, Silveira VS, Che J, Corcoran RB, Bardeesy N. Landscape of Clinical Resistance Mechanisms to FGFR Inhibitors in FGFR2-Altered Cholangiocarcinoma. Clin Cancer Res 2024; 30:198-208. [PMID: 37843855 PMCID: PMC10767308 DOI: 10.1158/1078-0432.ccr-23-1317] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/18/2023] [Accepted: 10/12/2023] [Indexed: 10/17/2023]
Abstract
PURPOSE FGFR inhibitors are effective in FGFR2-altered cholangiocarcinoma, leading to approval of reversible FGFR inhibitors, pemigatinib and infigratinib, and an irreversible inhibitor, futibatinib. However, acquired resistance develops, limiting clinical benefit. Some mechanisms of resistance have been reported, including secondary FGFR2 kinase domain mutations. Here, we sought to establish the landscape of acquired resistance to FGFR inhibition and to validate findings in model systems. EXPERIMENTAL DESIGN We examined the spectrum of acquired resistance mechanisms detected in circulating tumor DNA or tumor tissue upon disease progression following FGFR inhibitor therapy in 82 FGFR2-altered cholangiocarcinoma patients from 12 published reports. Functional studies of candidate resistance alterations were performed. RESULTS Overall, 49 of 82 patients (60%) had one or more detectable secondary FGFR2 kinase domain mutations upon acquired resistance. N550 molecular brake and V565 gatekeeper mutations were most common, representing 63% and 47% of all FGFR2 kinase domain mutations, respectively. Functional studies showed different inhibitors displayed unique activity profiles against FGFR2 mutations. Interestingly, disruption of the cysteine residue covalently bound by futibatinib (FGFR2 C492) was rare, observed in 1 of 42 patients treated with this drug. FGFR2 C492 mutations were insensitive to inhibition by futibatinib but showed reduced signaling activity, potentially explaining their low frequency. CONCLUSIONS These data support secondary FGFR2 kinase domain mutations as the primary mode of acquired resistance to FGFR inhibitors, most commonly N550 and V565 mutations. Thus, development of combination strategies and next-generation FGFR inhibitors targeting the full spectrum of FGFR2 resistance mutations will be critical.
Collapse
Affiliation(s)
- Qibiao Wu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Haley Ellis
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Giulia Siravegna
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Alexa G. Michel
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Bryanna L. Norden
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Ferran Fece de la Cruz
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Eranga Roshan Balasooriya
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Yuanli Zhen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Vanessa S. Silveira
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Jianwe Che
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ryan B. Corcoran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| |
Collapse
|
30
|
Wu Z, Huang D, Wang J, Zhao Y, Sun W, Shen X. Engineering Heterogeneous Tumor Models for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304160. [PMID: 37946674 PMCID: PMC10767453 DOI: 10.1002/advs.202304160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/16/2023] [Indexed: 11/12/2023]
Abstract
Tumor tissue engineering holds great promise for replicating the physiological and behavioral characteristics of tumors in vitro. Advances in this field have led to new opportunities for studying the tumor microenvironment and exploring potential anti-cancer therapeutics. However, the main obstacle to the widespread adoption of tumor models is the poor understanding and insufficient reconstruction of tumor heterogeneity. In this review, the current progress of engineering heterogeneous tumor models is discussed. First, the major components of tumor heterogeneity are summarized, which encompasses various signaling pathways, cell proliferations, and spatial configurations. Then, contemporary approaches are elucidated in tumor engineering that are guided by fundamental principles of tumor biology, and the potential of a bottom-up approach in tumor engineering is highlighted. Additionally, the characterization approaches and biomedical applications of tumor models are discussed, emphasizing the significant role of engineered tumor models in scientific research and clinical trials. Lastly, the challenges of heterogeneous tumor models in promoting oncology research and tumor therapy are described and key directions for future research are provided.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Danqing Huang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Jinglin Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| | - Weijian Sun
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Xian Shen
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| |
Collapse
|
31
|
Guo T, Wang J, Pang M, Liu W, Zhang X, Fan A, Liu H, Liu Q, Wei T, Li C, Zhao X, Lu Y. Reprogramming and multi-lineage transdifferentiation attenuate the tumorigenicity of colorectal cancer cells. J Biol Chem 2024; 300:105534. [PMID: 38072050 PMCID: PMC10801221 DOI: 10.1016/j.jbc.2023.105534] [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: 05/29/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 01/02/2024] Open
Abstract
Significant advances have been made in reprogramming various somatic cells into induced pluripotent stem cells (iPSCs) and in multi-lineage differentiation (transdifferentiation) into different tissues. These manipulable transdifferentiating techniques may be applied in cancer therapy. Limited works have been reported that cancer cell malignancy can be switched to benign phenotypes through reprogramming techniques. Here, we reported that two colorectal cancer (CRC) cell lines (DLD1, HT29) could be reprogrammed into iPSCs (D-iPSCs, H-iPSCs). D- and H-iPSCs showed reduced tumorigenesis. Furthermore, we successfully induced D- and H-iPSCs differentiation into terminally differentiated cell types such as cardiomyocyte, neuron, and adipocyte-like cells. Impressively, the differentiated cells exhibited further attenuated tumorigenesis in vitro and in vivo. RNA-Seq further indicated that epigenetic changes occurred after reprogramming and transdifferentiation that caused reduced tumorigenicity. Overall, our study indicated that CRC cells can be reprogrammed and further differentiated into terminally differentiated lineages with attenuation of their malignancy in vitro and in vivo. The current work sheds light on a potential multi-lineage differentiation therapeutic strategy for colorectal cancer.
Collapse
Affiliation(s)
- Tongtong Guo
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Juan Wang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Maogui Pang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China; Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wanning Liu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaohui Zhang
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ahui Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hengtao Liu
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China
| | - Qianqian Liu
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China
| | - Tianying Wei
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China
| | - Cunxi Li
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China; Cytogenetics Laboratory, Beijing Institute of Human Genetics and Reproduction Medicine, Beijing, China.
| | - Xiaodi Zhao
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Yuanyuan Lu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
32
|
Williams CJ, Elliott F, Sapanara N, Aghaei F, Zhang L, Muranyi A, Yan D, Bai I, Zhao Z, Shires M, Wood HM, Richman SD, Hemmings G, Hale M, Bottomley D, Galvin L, Cartlidge C, Dance S, Bacon CM, Mansfield L, Young-Zvandasara K, Sudan A, Lambert K, Bibby I, Coupland SE, Montazeri A, Kipling N, Hughes K, Cross SS, Dewdney A, Pheasey L, Leng C, Gochera T, Mangham DC, Saunders M, Pritchard M, Stott H, Mukherjee A, Ilyas M, Silverman R, Hyland G, Sculthorpe D, Thornton K, Gould I, O'Callaghan A, Brown N, Turnbull S, Shaw L, Seymour MT, West NP, Seligmann JF, Singh S, Shanmugam K, Quirke P. Associations between AI-Assisted Tumor Amphiregulin and Epiregulin IHC and Outcomes from Anti-EGFR Therapy in the Routine Management of Metastatic Colorectal Cancer. Clin Cancer Res 2023; 29:4153-4165. [PMID: 37363997 PMCID: PMC10570673 DOI: 10.1158/1078-0432.ccr-23-0859] [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: 03/24/2023] [Revised: 05/31/2023] [Accepted: 06/22/2023] [Indexed: 06/28/2023]
Abstract
PURPOSE High tumor production of the EGFR ligands, amphiregulin (AREG) and epiregulin (EREG), predicted benefit from anti-EGFR therapy for metastatic colorectal cancer (mCRC) in a retrospective analysis of clinical trial data. Here, AREG/EREG IHC was analyzed in a cohort of patients who received anti-EGFR therapy as part of routine care, including key clinical contexts not investigated in the previous analysis. EXPERIMENTAL DESIGN Patients who received panitumumab or cetuximab ± chemotherapy for treatment of RAS wild-type mCRC at eight UK cancer centers were eligible. Archival formalin-fixed paraffin-embedded tumor tissue was analyzed for AREG and EREG IHC in six regional laboratories using previously developed artificial intelligence technologies. Primary endpoints were progression-free survival (PFS) and overall survival (OS). RESULTS A total of 494 of 541 patients (91.3%) had adequate tissue for analysis. A total of 45 were excluded after central extended RAS testing, leaving 449 patients in the primary analysis population. After adjustment for additional prognostic factors, high AREG/EREG expression (n = 360; 80.2%) was associated with significantly prolonged PFS [median: 8.5 vs. 4.4 months; HR, 0.73; 95% confidence interval (CI), 0.56-0.95; P = 0.02] and OS [median: 16.4 vs. 8.9 months; HR, 0.66 95% CI, 0.50-0.86; P = 0.002]. The significant OS benefit was maintained among patients with right primary tumor location (PTL), those receiving cetuximab or panitumumab, those with an oxaliplatin- or irinotecan-based chemotherapy backbone, and those with tumor tissue obtained by biopsy or surgical resection. CONCLUSIONS High tumor AREG/EREG expression was associated with superior survival outcomes from anti-EGFR therapy in mCRC, including in right PTL disease. AREG/EREG IHC assessment could aid therapeutic decisions in routine practice. See related commentary by Randon and Pietrantonio, p. 4021.
Collapse
Affiliation(s)
- Christopher J.M. Williams
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Faye Elliott
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Nancy Sapanara
- Medical & Scientific Affairs, Roche Molecular Systems Inc., Tucson, Arizona
| | - Faranak Aghaei
- Medical & Scientific Affairs, Roche Molecular Systems Inc., Tucson, Arizona
| | - Liping Zhang
- Medical & Scientific Affairs, Roche Molecular Systems Inc., Tucson, Arizona
| | - Andrea Muranyi
- Medical & Scientific Affairs, Roche Molecular Systems Inc., Tucson, Arizona
| | - Dongyao Yan
- Medical & Scientific Affairs, Roche Molecular Systems Inc., Tucson, Arizona
| | - Isaac Bai
- Medical & Scientific Affairs, Roche Molecular Systems Inc., Tucson, Arizona
| | - Zuo Zhao
- Imaging and Algorithms, Digital Pathology, Roche Sequencing Solutions Inc., Santa Clara, California
| | - Michael Shires
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Henry M. Wood
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Susan D. Richman
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Gemma Hemmings
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Michael Hale
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Daniel Bottomley
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Leanne Galvin
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Caroline Cartlidge
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Sarah Dance
- Medical Affairs, Access and Innovation, Roche Diagnostics Limited, Burgess Hill, United Kingdom
| | - Chris M. Bacon
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Laura Mansfield
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | | | - Ajay Sudan
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Katy Lambert
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Irena Bibby
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Sarah E. Coupland
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Amir Montazeri
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, United Kingdom
| | - Natalie Kipling
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Kathryn Hughes
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, United Kingdom
| | - Simon S. Cross
- Academic Unit of Pathology, Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Alice Dewdney
- Weston Park Cancer Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Leanne Pheasey
- Weston Park Cancer Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Cathryn Leng
- Weston Park Cancer Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Tatenda Gochera
- Weston Park Cancer Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - D. Chas Mangham
- Adult Histopathology, Laboratory Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, United Kingdom
| | - Mark Saunders
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Martin Pritchard
- Adult Histopathology, Laboratory Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, United Kingdom
| | - Helen Stott
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Abhik Mukherjee
- Translational Medical Sciences, Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Mohammad Ilyas
- Translational Medical Sciences, Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Rafael Silverman
- Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Georgina Hyland
- Translational Medical Sciences, Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Declan Sculthorpe
- Translational Medical Sciences, Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Kirsty Thornton
- Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Imogen Gould
- Translational Medical Sciences, Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | | | - Nicholas Brown
- Calderdale and Huddersfield NHS Foundation Trust, Huddersfield, United Kingdom
| | - Samantha Turnbull
- Calderdale and Huddersfield NHS Foundation Trust, Huddersfield, United Kingdom
| | - Lisa Shaw
- Calderdale and Huddersfield NHS Foundation Trust, Huddersfield, United Kingdom
| | - Matthew T. Seymour
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Nicholas P. West
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Jenny F. Seligmann
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Shalini Singh
- Medical & Scientific Affairs, Roche Molecular Systems Inc., Tucson, Arizona
| | - Kandavel Shanmugam
- Medical & Scientific Affairs, Roche Molecular Systems Inc., Tucson, Arizona
| | - Philip Quirke
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
33
|
Safaei M, Khalighi F, Behabadi FA, Abpeikar Z, Goodarzi A, Kouhpayeh SA, Najafipour S, Ramezani V. Liposomal nanocarriers containing siRNA as small molecule-based drugs to overcome cancer drug resistance. Nanomedicine (Lond) 2023; 18:1745-1768. [PMID: 37965906 DOI: 10.2217/nnm-2023-0176] [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: 11/16/2023] Open
Abstract
This review discusses the application of nanoliposomes containing siRNA/drug to overcome multidrug resistance for all types of cancer treatments. As drug resistance-associated factors are overexpressed in many cancer cell types, pumping chemotherapy drugs out of the cytoplasm leads to an inadequate therapeutic response. The siRNA/drug-loaded nanoliposomes are a promising approach to treating multidrug-resistant cancer, as they can effectively transmit a small-molecule drug into the target cytoplasm, ensuring that the drug binds efficiently. Moreover, nanoliposome-based therapeutics with advances in nanotechnology can effectively deliver siRNA to cancer cells. Overall, nanoliposomes have the potential to effectively deliver siRNA and small-molecule drugs in a targeted manner and are thus a promising tool for the treatment of cancer and other diseases.
Collapse
Affiliation(s)
- Mohsen Safaei
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, 7461686688, Iran
| | - Fatemeh Khalighi
- Department of Pharmaceutics, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, 9417694780, Iran
| | - Fatemeh Akhavan Behabadi
- Department of Pharmaceutics, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, 9417694780, Iran
| | - Zahra Abpeikar
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, 7461686688, Iran
| | - Arash Goodarzi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, 7461686688, Iran
| | - Seyed Amin Kouhpayeh
- Department of Pharmacology, School of Medicine, Fasa University of Medical Sciences, Fasa, 7461686688, Iran
| | - Sohrab Najafipour
- Department of Microbiology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, 7461686688, Iran
| | - Vahid Ramezani
- Department of Pharmaceutics, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, 9417694780, Iran
- Pharmaceutical Sciences Research Center, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, 9417694780, Iran
| |
Collapse
|
34
|
Tsai HL, Lin CC, Sung YC, Chen SH, Chen LT, Jiang JK, Wang JY. The emergence of RAS mutations in patients with RAS wild-type mCRC receiving cetuximab as first-line treatment: a noninterventional, uncontrolled multicenter study. Br J Cancer 2023; 129:947-955. [PMID: 37488448 PMCID: PMC10491612 DOI: 10.1038/s41416-023-02366-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023] Open
Abstract
ABSRTACT BACKGROUND: Patients treated with anti-epidermal growth factor receptor (anti-EGFR) will ultimately develop acquired resistance promoted by clonal selection, mainly the emergence of mutations in the MAPK pathway (mostly RAS mutations). Baseline assessment of RAS mutations in the blood of patients correlates well with RAS tumour tissue testing and is currently an alternative option in routine clinical practice to guide first-line therapy. The aim of this study was the prevalence of acquired genomic alterations detected in the auxiliary tool of ctDNA testing and investigated the role of RAS ctDNA status for detecting tumour response and predicting benefit to anti-EGFR therapy. METHODS Only patients with concordant wild-type formalin-fixed, paraffin-embedded (FFPE) tumour tissue and baseline ctDNA RAS wild-type were included. RAS mutations in plasma were evaluated using MassARRAY platform. Blood samples were collected at baseline, every 3 months during first-line treatment, and at disease progression. The primary endpoint was the detection rate of RAS mutations during cetuximab treatment. The correlation between response and survival outcomes and the emergence of circulating RAS mutations was also analysed. RESULTS The detection rate of RAS mutations during treatment was 9.3% (10/108). RAS mutations detection occurred a median of 3 months prior to radiologic documentation. The subgroup of patients with RAS mutations exhibited significantly inferior progression-free survival and overall survival (P = 0.002 and 0.027, respectively) but the baseline characteristics, response rates, disease control rates, and metastatectomy were not significant (all P > 0.05). CONCLUSIONS We demonstrated that RAS ctDNA status might be a valuable biomarker for detecting early tumour response and predicting benefit to anti-EGFR therapy. CLINICAL TRIAL REGISTRATION NCT03401957 (January 17, 2018).
Collapse
Affiliation(s)
- Hsiang-Lin Tsai
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Chi Lin
- Division of Colorectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yung-Chung Sung
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
- Division of Hematology/Oncology, Internal Medicine, Cathay General Hospital, Taipei, Taiwan
| | - Shang-Hung Chen
- Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Division of Medical Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jeng-Kai Jiang
- Division of Colorectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Jaw-Yuan Wang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Pingtung Hospital, Ministry of Health and Welfare, Pingtung, Taiwan.
| |
Collapse
|
35
|
Altintas DM, Comoglio PM. An Observatory for the MET Oncogene: A Guide for Targeted Therapies. Cancers (Basel) 2023; 15:4672. [PMID: 37760640 PMCID: PMC10526818 DOI: 10.3390/cancers15184672] [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: 08/31/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
The MET proto-oncogene encodes a pivotal tyrosine kinase receptor, binding the hepatocyte growth factor (HGF, also known as scatter factor, SF) and governing essential biological processes such as organogenesis, tissue repair, and angiogenesis. The pleiotropic physiological functions of MET explain its diverse role in cancer progression in a broad range of tumors; genetic/epigenetic alterations of MET drive tumor cell dissemination, metastasis, and acquired resistance to conventional and targeted therapies. Therefore, targeting MET emerged as a promising strategy, and many efforts were devoted to identifying the optimal way of hampering MET signaling. Despite encouraging results, however, the complexity of MET's functions in oncogenesis yields intriguing observations, fostering a humbler stance on our comprehension. This review explores recent discoveries concerning MET alterations in cancer, elucidating their biological repercussions, discussing therapeutic avenues, and outlining future directions. By contextualizing the research question and articulating the study's purpose, this work navigates MET biology's intricacies in cancer, offering a comprehensive perspective.
Collapse
Affiliation(s)
| | - Paolo M. Comoglio
- IFOM ETS—The AIRC Institute of Molecular Oncology, 20139 Milano, Italy;
| |
Collapse
|
36
|
Lu N, Guan X, Zhu J, Li Y, Zhang J. A Contrast-Enhanced CT-Based Deep Learning System for Preoperative Prediction of Colorectal Cancer Staging and RAS Mutation. Cancers (Basel) 2023; 15:4497. [PMID: 37760468 PMCID: PMC10526233 DOI: 10.3390/cancers15184497] [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: 08/23/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
PURPOSE This study aimed to build a deep learning system using enhanced computed tomography (CT) portal-phase images for predicting colorectal cancer patients' preoperative staging and RAS gene mutation status. METHODS The contrast-enhanced CT image dataset comprises the CT portal-phase images from a retrospective cohort of 231 colorectal cancer patients. The deep learning system was developed via migration learning for colorectal cancer detection, staging, and RAS gene mutation status prediction. This study used pre-trained Yolov7, vision transformer (VIT), swin transformer (SWT), EfficientNetV2, and ConvNeXt. 4620, and contrast-enhanced CT images and annotated tumor bounding boxes were included in the tumor identification and staging dataset. A total of 19,700 contrast-enhanced CT images comprise the RAS gene mutation status prediction dataset. RESULTS In the validation cohort, the Yolov7-based detection model detected and staged tumors with a mean accuracy precision (IoU = 0.5) (mAP_0.5) of 0.98. The area under the receiver operating characteristic curve (AUC) in the test set and validation set for the VIT-based prediction model in predicting the mutation status of the RAS genes was 0.9591 and 0.9554, respectively. The detection network and prediction network of the deep learning system demonstrated great performance in explaining contrast-enhanced CT images. CONCLUSION In this study, a deep learning system was created based on the foundation of contrast-enhanced CT portal-phase imaging to preoperatively predict the stage and RAS mutation status of colorectal cancer patients. This system will help clinicians choose the best treatment option to increase colorectal cancer patients' chances of survival and quality of life.
Collapse
Affiliation(s)
- Na Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No. 121, Jiangjiayuan Road, Nanjing 210011, China (X.G.)
| | - Xiao Guan
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No. 121, Jiangjiayuan Road, Nanjing 210011, China (X.G.)
| | - Jianguo Zhu
- Department of Radiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China;
| | - Yuan Li
- Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China;
| | - Jianping Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No. 121, Jiangjiayuan Road, Nanjing 210011, China (X.G.)
| |
Collapse
|
37
|
Hiltbrunner S, Cords L, Kasser S, Freiberger SN, Kreutzer S, Toussaint NC, Grob L, Opitz I, Messerli M, Zoche M, Soltermann A, Rechsteiner M, van den Broek M, Bodenmiller B, Curioni-Fontecedro A. Acquired resistance to anti-PD1 therapy in patients with NSCLC associates with immunosuppressive T cell phenotype. Nat Commun 2023; 14:5154. [PMID: 37620318 PMCID: PMC10449840 DOI: 10.1038/s41467-023-40745-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
Immune checkpoint inhibitor treatment has the potential to prolong survival in non-small cell lung cancer (NSCLC), however, some of the patients develop resistance following initial response. Here, we analyze the immune phenotype of matching tumor samples from a cohort of NSCLC patients showing good initial response to immune checkpoint inhibitors, followed by acquired resistance at later time points. By using imaging mass cytometry and whole exome and RNA sequencing, we detect two patterns of resistance¨: One group of patients is characterized by reduced numbers of tumor-infiltrating CD8+ T cells and reduced expression of PD-L1 after development of resistance, whereas the other group shows high CD8+ T cell infiltration and high expression of PD-L1 in addition to markedly elevated expression of other immune-inhibitory molecules. In two cases, we detect downregulation of type I and II IFN pathways following progression to resistance, which could lead to an impaired anti-tumor immune response. This study thus captures the development of immune checkpoint inhibitor resistance as it progresses and deepens our mechanistic understanding of immunotherapy response in NSCLC.
Collapse
Affiliation(s)
- Stefanie Hiltbrunner
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, 8091, Switzerland
- Comprehensive Cancer Center Zurich, University Hospital Zurich, Zurich, 8091, Switzerland
- University of Zurich, Zurich, Switzerland
- University of Fribourg, Faculty of Science and Medicine, Fribourg, 1700, Switzerland
| | - Lena Cords
- University of Zurich, Zurich, Switzerland
- Department of Quantitative Biomedicine, University of Zurich, Zurich, 8057, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, 8049, Switzerland
- Life Science Zurich Graduate School, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Sabrina Kasser
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, 8091, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Sandra N Freiberger
- Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Susanne Kreutzer
- Functional Genomics Center Zurich, ETH and University of Zurich, Zurich, 8057, Switzerland
| | - Nora C Toussaint
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, 8952, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Linda Grob
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, 8952, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, 8091, Switzerland
| | - Michael Messerli
- University of Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, 8091, Switzerland
| | - Martin Zoche
- Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Alex Soltermann
- Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091, Zurich, Switzerland
- Pathologie Länggasse, Ittigen, 3063, Switzerland
| | - Markus Rechsteiner
- Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Maries van den Broek
- University of Zurich, Zurich, Switzerland
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Bernd Bodenmiller
- University of Zurich, Zurich, Switzerland
- Department of Quantitative Biomedicine, University of Zurich, Zurich, 8057, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, 8049, Switzerland
| | - Alessandra Curioni-Fontecedro
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, 8091, Switzerland.
- Comprehensive Cancer Center Zurich, University Hospital Zurich, Zurich, 8091, Switzerland.
- University of Zurich, Zurich, Switzerland.
- University of Fribourg, Faculty of Science and Medicine, Fribourg, 1700, Switzerland.
- Clinic of Oncology, Cantonal Hospital Fribourg, Fribourg, 1752, Switzerland.
| |
Collapse
|
38
|
Qi Z, Li Y, Wang Z, Tan X, Zhou Y, Li Z, Zhao W, Zheng X, Yao J, Li F, Wang W, Wang Z, Pang F, Wang G, Gu W. Monitoring of gastrointestinal carcinoma via molecular residual disease with circulating tumor DNA using a tumor-informed assay. Cancer Med 2023; 12:16687-16696. [PMID: 37602656 PMCID: PMC10501225 DOI: 10.1002/cam4.6286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 02/02/2023] [Accepted: 06/14/2023] [Indexed: 08/22/2023] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA)-based minimal residual disease (MRD) detection, which can identify disease relapse ahead of radiological imaging, has shown promising performance. The objective of this study was to develop and validate OriMIRACLE S (Minimal Residual Circulating Nucleic Acid Longitudinal Detection in Solid Tumor), a highly sensitive and specific tumor-informed assay for MRD detection. METHODS Tumor-specific somatic single nucleotide variants (SNVs) were identified via whole exome sequencing of tumor tissue and matched germline DNA. Clonal SNVs were selected using the OriSelector algorithm for patient-specific, multiplex PCR-based NGS assays in MRD detection. Plasma-free DNA from patients with gastrointestinal tumors prior to and following an operation, and during monitoring, were ultradeep sequenced. RESULTS The detection of three positive sites was sufficient to achieve nearly 100% overall sample level sensitivity and specificity and was determined by calculating binomial probability based on customized panels containing 21 to 30 variants. A total of 127 patients with gastrointestinal tumors were enrolled in our study. Preoperatively, MRD was positive in 18 of 26 patients (69.23%). Following surgery, MRD was positive in 24 of 82 patients (29.27%). The positivity rate for MRD was 33.33% (n = 18) for gastric adenocarcinoma and 32.26% (n = 62) for colorectal cancer. Twenty (20) of 59 patients (34.48%) experienced a change in MRD status over the monitoring period. Patients 8 and 31 responded to 3 cycles of systemic therapy, after which levels for all ctDNA dropped below the detection limit. Patient 53 was an example of using MRD to predict tumor metastasis. Patient 55 showed a weak response to treatments first and respond to new systemic therapy after tumor progression. CONCLUSION Our study identified a sensitive and specific clinical detection method for low frequency ctDNA, and explored the detection performance of this technology in gastrointestinal tumors.
Collapse
Affiliation(s)
- Zining Qi
- Department of Gastrointestinal surgeryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Yi Li
- Department of Gastrointestinal surgeryThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - ZhengKun Wang
- Department of Gastrointestinal surgeryThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Xuerong Tan
- Department of RadiologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Yixuan Zhou
- Department of RadiologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | | | | | - Xin Zheng
- Shanghai OrigiMed Co., LtdShanghaiChina
| | | | - Feng Li
- Shanghai OrigiMed Co., LtdShanghaiChina
| | | | | | - Fei Pang
- Shanghai OrigiMed Co., LtdShanghaiChina
| | - Gang Wang
- Department of General SurgeryJiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Weiguang Gu
- Department of oncologyNanhai People's HospitalFoshanChina
- Department of The Sixth Affiliated HospitalSchool of Medicine, South China University of TechnologyFoshanChina
| |
Collapse
|
39
|
Powell CL, Saddoughi SA, Wigle DA. Progress in genome-inspired treatment decisions for multifocal lung adenocarcinoma. Expert Rev Respir Med 2023; 17:1009-1021. [PMID: 37982734 DOI: 10.1080/17476348.2023.2286277] [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: 07/05/2023] [Accepted: 11/17/2023] [Indexed: 11/21/2023]
Abstract
INTRODUCTION Multifocal lung adenocarcinoma (MFLA) is becoming increasingly recognized as a distinct subset of lung cancer, with unique biology, disease course, and treatment outcomes. While definitions remain controversial, MFLA is characterized by the development and concurrent presence of multiple independent (non-metastatic) lesions on the lung adenocarcinoma spectrum. Disease progression typically follows an indolent course measured in years, with a lower propensity for nodal and distant metastases than other more common forms of non-small cell lung cancer. AREAS COVERED Traditional imaging and histopathological analyses of tumor biopsies are frequently unable to fully characterize the disease, prompting interest in molecular diagnosis. We highlight some of the key questions in the field, including accurate definitions to identify and stage MLFA, molecular tests to stratify patients and treatment decisions, and the lack of clinical trial data to delineate best management for this poorly understood subset of lung cancer patients. We review the existing literature and progress toward a genomic diagnosis for this unique disease entity. EXPERT OPINION Multifocal lung adenocarcinoma behaves differently than other forms of non-small cell lung cancer. Progress in molecular diagnosis may enhance potential for accurate definition, diagnosis, and optimizing treatment approach.
Collapse
Affiliation(s)
- Chelsea L Powell
- Division of Thoracic Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Sahar A Saddoughi
- Division of Thoracic Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Dennis A Wigle
- Division of Thoracic Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
40
|
Nakamura R, Fujii H, Yamada T, Matsui Y, Yaoi T, Honda M, Tanaka N, Miyagawa-Hayashino A, Yoshimura A, Morimoto K, Iwasaku M, Tokuda S, Kim YH, Konishi E, Itoh K, Takayama K. Analysis of Tumor Heterogeneity Through AXL Activation in Primary Resistance to EGFR Tyrosine Kinase Inhibitors. JTO Clin Res Rep 2023; 4:100525. [PMID: 37426308 PMCID: PMC10329144 DOI: 10.1016/j.jtocrr.2023.100525] [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: 12/22/2022] [Revised: 04/24/2023] [Accepted: 05/08/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction EGFR tyrosine kinase inhibitors are standard therapeutic agents for patients with advanced NSCLC harboring EGFR mutations. Nevertheless, some patients exhibit primary resistance to EGFR tyrosine kinase inhibitors in the first-line treatment setting. AXL, a member of the TYRO3, AXL, and MERTK family of receptor tyrosine kinases, is involved in primary resistance to EGFR tyrosine kinase inhibitors in EGFR-mutated NSCLC. Methods We investigated spatial tumor heterogeneity using autopsy specimens and a patient-derived cell line from a patient with EGFR-mutated NSCLC having primary resistance to erlotinib plus ramucirumab. Results Quantitative polymerase chain reaction analysis revealed that AXL mRNA expression differed at each metastatic site. In addition, AXL expression levels were likely to be negatively correlated with the effectiveness of erlotinib plus ramucirumab therapy. Analysis of a patient-derived cell line established from the left pleural effusion before initiation of treatment revealed that the combination of EGFR tyrosine kinase inhibitors and an AXL inhibitor remarkably inhibited cell viability and increased cell apoptosis in comparison with EGFR tyrosine kinase inhibitor monotherapy or combination therapy of these inhibitors with ramucirumab. Conclusions Our observations suggest that AXL expression may play a critical role in the progression of spatial tumor heterogeneity and primary resistance to EGFR tyrosine kinase inhibitors in patients with EGFR-mutated NSCLC.
Collapse
Affiliation(s)
- Ryota Nakamura
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyuki Fujii
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tadaaki Yamada
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yohei Matsui
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Yaoi
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mizuki Honda
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Noriyuki Tanaka
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Aya Miyagawa-Hayashino
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akihiro Yoshimura
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenji Morimoto
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masahiro Iwasaku
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shinsaku Tokuda
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Young Hak Kim
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiichi Konishi
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
41
|
Liu QL, Zhou H, Zhou ZG, Chen HN. Colorectal cancer liver metastasis: genomic evolution and crosstalk with the liver microenvironment. Cancer Metastasis Rev 2023; 42:575-587. [PMID: 37061644 DOI: 10.1007/s10555-023-10107-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/10/2023] [Indexed: 04/17/2023]
Abstract
Colorectal cancer (CRC) patients frequently develop liver metastases, which are the major cause of cancer-related mortality. The molecular basis and management of colorectal liver metastases (CRLMs) remain a challenging clinical issue. Recent genomic evidence has demonstrated the liver tropism of CRC and the presence of a stricter evolutionary bottleneck in the liver as a target organ compared to lymph nodes. This bottleneck challenging CRC cells in the liver is organ-specific and requires adaptation not only at the genetic level, but also at the phenotypic level to crosstalk with the hepatic microenvironment. Here, we highlight the emerging evidence on the clonal evolution of CRLM and review recent insights into the molecular mechanisms orchestrating the bidirectional interactions between metastatic CRC cells and the unique liver microenvironment.
Collapse
Affiliation(s)
- Qiu-Luo Liu
- Department of General Surgery, Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Huijie Zhou
- Department of Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zong-Guang Zhou
- Department of General Surgery, Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Hai-Ning Chen
- Department of General Surgery, Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
42
|
Inchingolo R, Maino C, Cannella R, Vernuccio F, Cortese F, Dezio M, Pisani AR, Giandola T, Gatti M, Giannini V, Ippolito D, Faletti R. Radiomics in colorectal cancer patients. World J Gastroenterol 2023; 29:2888-2904. [PMID: 37274803 PMCID: PMC10237092 DOI: 10.3748/wjg.v29.i19.2888] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/07/2023] [Accepted: 04/25/2023] [Indexed: 05/16/2023] Open
Abstract
The main therapeutic options for colorectal cancer are surgical resection and adjuvant chemotherapy in non-metastatic disease. However, the evaluation of the overall adjuvant chemotherapy benefit in patients with a high risk of recurrence is challenging. Radiological images can represent a source of data that can be analyzed by using automated computer-based techniques, working on numerical information coded within Digital Imaging and Communications in Medicine files: This image numerical analysis has been named "radiomics". Radiomics allows the extraction of quantitative features from radiological images, mainly invisible to the naked eye, that can be further analyzed by artificial intelligence algorithms. Radiomics is expanding in oncology to either understand tumor biology or for the development of imaging biomarkers for diagnosis, staging, and prognosis, prediction of treatment response and diseases monitoring and surveillance. Several efforts have been made to develop radiomics signatures for colorectal cancer patient using computed tomography (CT) images with different aims: The preoperative prediction of lymph node metastasis, detecting BRAF and RAS gene mutations. Moreover, the use of delta-radiomics allows the analysis of variations of the radiomics parameters extracted from CT scans performed at different timepoints. Most published studies concerning radiomics and magnetic resonance imaging (MRI) mainly focused on the response of advanced tumors that underwent neoadjuvant therapy. Nodes status is the main determinant of adjuvant chemotherapy. Therefore, several radiomics model based on MRI, especially on T2-weighted images and ADC maps, for the preoperative prediction of nodes metastasis in rectal cancer has been developed. Current studies mostly focused on the applications of radiomics in positron emission tomography/CT for the prediction of survival after curative surgical resection and assessment of response following neoadjuvant chemoradiotherapy. Since colorectal liver metastases develop in about 25% of patients with colorectal carcinoma, the main diagnostic tasks of radiomics should be the detection of synchronous and metachronous lesions. Radiomics could be an additional tool in clinical setting, especially in identifying patients with high-risk disease. Nevertheless, radiomics has numerous shortcomings that make daily use extremely difficult. Further studies are needed to assess performance of radiomics in stratifying patients with high-risk disease.
Collapse
Affiliation(s)
- Riccardo Inchingolo
- Unit of Interventional Radiology, F. Miulli Hospital, Acquaviva delle Fonti 70021, Italy
| | - Cesare Maino
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy
| | - Roberto Cannella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo 90127, Italy
| | - Federica Vernuccio
- Institute of Radiology, University Hospital of Padova, Padova 35128, Italy
| | - Francesco Cortese
- Unit of Interventional Radiology, F. Miulli Hospital, Acquaviva delle Fonti 70021, Italy
| | - Michele Dezio
- Unit of Interventional Radiology, F. Miulli Hospital, Acquaviva delle Fonti 70021, Italy
| | - Antonio Rosario Pisani
- Interdisciplinary Department of Medicine, Section of Nuclear Medicine, University of Bari “Aldo Moro”, Bari 70121, Italy
| | - Teresa Giandola
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy
| | - Marco Gatti
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Valentina Giannini
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Davide Ippolito
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy
| | - Riccardo Faletti
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| |
Collapse
|
43
|
Cai M, Song XL, Li XA, Chen M, Guo J, Yang DH, Chen Z, Zhao SC. Current therapy and drug resistance in metastatic castration-resistant prostate cancer. Drug Resist Updat 2023; 68:100962. [PMID: 37068396 DOI: 10.1016/j.drup.2023.100962] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/06/2023] [Accepted: 04/10/2023] [Indexed: 04/19/2023]
Abstract
Castration-resistant prostate cancer (CRPC), especially metastatic castration-resistant prostate cancer (mCRPC) is one of the most prevalent malignancies and main cause of cancer-related death among men in the world. In addition, it is very difficult for clinical treatment because of the natural or acquired drug resistance of CRPC. Mechanisms of drug resistance are extremely complicated and how to overcome it remains an urgent clinical problem to be solved. Thus, a comprehensive and thorough understanding for mechanisms of drug resistance in mCRPC is indispensable to develop novel and better therapeutic strategies. In this review, we aim to review new insight of the treatment of mCRPC and elucidate mechanisms governing resistance to new drugs: taxanes, androgen receptor signaling inhibitors (ARSIs) and poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi). Most importantly, in order to improve efficacy of these drugs, strategies of overcoming drug resistance are also discussed based on their mechanisms respectively.
Collapse
Affiliation(s)
- Maoping Cai
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong, PR China; The Third Clinical College, Southern Medical University, Guangzhou 510630, Guangdong, PR China; Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524045, Guangdong, PR China
| | - Xian-Lu Song
- Department of Radiotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, PR China
| | - Xin-An Li
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, PR China
| | - Mingkun Chen
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong, PR China; The Third Clinical College, Southern Medical University, Guangzhou 510630, Guangdong, PR China; Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, PR China
| | - Jiading Guo
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong, PR China; The Third Clinical College, Southern Medical University, Guangzhou 510630, Guangdong, PR China
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, Mineola 11501, NY, USA.
| | - Zhanghui Chen
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524045, Guangdong, PR China.
| | - Shan-Chao Zhao
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong, PR China; The Third Clinical College, Southern Medical University, Guangzhou 510630, Guangdong, PR China; Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, PR China.
| |
Collapse
|
44
|
Yokoyama S, Watanabe T, Fujita Y, Matsumura S, Ueda K, Nagano S, Kinoshita I, Murakami D, Tabata H, Tsuji T, Ozawa S, Tamaki T, Nakatani Y, Oka M. Histology of metastatic colorectal cancer in a lymph node. PLoS One 2023; 18:e0284536. [PMID: 37053292 PMCID: PMC10101456 DOI: 10.1371/journal.pone.0284536] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/31/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND A primary colorectal cancer (CRC) tumor can contain heterogeneous cancer cells. As clones of cells with different properties metastasize to lymph nodes (LNs), they could show different morphologies. Cancer histologies in LNs of CRC remains to be described. METHODS Our study enrolled 318 consecutive patients with CRC who underwent primary tumor resection with lymph node dissection between January 2011 and June 2016. 119 (37.4%) patients who had metastatic LNs (mLNs) were finally included in this study. Cancer histologies in LNs were classified and compared with pathologically diagnosed differentiation in the primary lesion. The association between histologies in lymph node metastasis (LNM) and prognosis in patients with CRC was investigated. RESULTS The histologies of the cancer cells in the mLNs were classified into four types: tubular, cribriform, poorly differentiated, and mucinous. Same degree of pathologically diagnosed differentiation in the primary tumor produced various histological types in LNM. In Kaplan-Meier analysis, prognosis was worse in CRC patients with moderately differentiated adenocarcinoma who had at least some mLN also showing cribriform carcinoma than for those whose mLNs all showed tubular carcinoma. CONCLUSIONS Histology in LNM from CRC might indicate the heterogeneity and malignant phenotype of the disease.
Collapse
Affiliation(s)
- Shozo Yokoyama
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Takashi Watanabe
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Yoichi Fujita
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Shuichi Matsumura
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Katsuya Ueda
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Shotaro Nagano
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Ikuharu Kinoshita
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Daisuke Murakami
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Hirotaka Tabata
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Toshiaki Tsuji
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Satoru Ozawa
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Takuya Tamaki
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Yoshihiro Nakatani
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| | - Masami Oka
- Department of Surgery, National Hospital Organization Minami Wakayama Medical Center, Tanabe, Japan
| |
Collapse
|
45
|
Khan SR, Scheffler M, Soomar SM, Rashid YA, Moosajee M, Ahmad A, Raza A, Uddin S. Role of circulating-tumor DNA in the early-stage non-small cell lung carcinoma as a predictive biomarker. Pathol Res Pract 2023; 245:154455. [PMID: 37054576 DOI: 10.1016/j.prp.2023.154455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/07/2023] [Accepted: 04/07/2023] [Indexed: 04/15/2023]
Abstract
Lung cancer is one of the most common solid malignancies. Tissue biopsy is the standard method for accurately diagnosing lung and many other malignancies over decades. However, molecular profiling of tumors leads to establishing a new horizon in the field of precision medicine, which has now entered the mainstream in clinical practice. In this context, a minimally invasive complementary method has been proposed as a liquid biopsy (LB) which is a blood-based test that is gaining popularity as it provides the opportunity to test genotypes in a unique, less invasive manner. Circulating tumor cells (CTC) captivating the Circulating-tumor DNA (Ct-DNA) are often present in the blood of lung cancer patients and are the fundamental concept behind LB. There are multiple clinical uses of Ct-DNA, including its role in prognostic and therapeutic purposes. The treatment of lung cancer has drastically evolved over time. Therefore, this review article mainly focuses on the current literature on circulating tumor DNA and its clinical implications and future goals in non-small cell lung cancer.
Collapse
Affiliation(s)
- Saqib Raza Khan
- Medical Oncology Department, Aga Khan University Hospital, Karachi, Pakistan.
| | - Matthias Scheffler
- Internal Medicine Department, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | | | - Yasmin Abdul Rashid
- Medical Oncology Department, Aga Khan University Hospital, Karachi, Pakistan
| | - Munira Moosajee
- Medical Oncology Department, Aga Khan University Hospital, Karachi, Pakistan
| | - Aamir Ahmad
- Translational Research Institute & Dermatology Institute, Hamad Medical Corporation, Doha, Qatar
| | - Afsheen Raza
- College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Shahab Uddin
- Translational Research Institute & Dermatology Institute, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
46
|
David P, Mittelstädt A, Kouhestani D, Anthuber A, Kahlert C, Sohn K, Weber GF. Current Applications of Liquid Biopsy in Gastrointestinal Cancer Disease-From Early Cancer Detection to Individualized Cancer Treatment. Cancers (Basel) 2023; 15:cancers15071924. [PMID: 37046585 PMCID: PMC10093361 DOI: 10.3390/cancers15071924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Worldwide, gastrointestinal (GI) cancers account for a significant amount of cancer-related mortality. Tests that allow an early diagnosis could lead to an improvement in patient survival. Liquid biopsies (LBs) due to their non-invasive nature as well as low risk are the current focus of cancer research and could be a promising tool for early cancer detection. LB involves the sampling of any biological fluid (e.g., blood, urine, saliva) to enrich and analyze the tumor's biological material. LBs can detect tumor-associated components such as circulating tumor DNA (ctDNA), extracellular vesicles (EVs), and circulating tumor cells (CTCs). These components can reflect the status of the disease and can facilitate clinical decisions. LBs offer a unique and new way to assess cancers at all stages of treatment, from cancer screenings to prognosis to management of multidisciplinary therapies. In this review, we will provide insights into the current status of the various types of LBs enabling early detection and monitoring of GI cancers and their use in in vitro diagnostics.
Collapse
Affiliation(s)
- Paul David
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anke Mittelstädt
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Dina Kouhestani
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anna Anthuber
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christoph Kahlert
- Department of Surgery, Carl Gustav Carus University Hospital, 01307 Dresden, Germany
| | - Kai Sohn
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
| | - Georg F Weber
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
47
|
Al Bitar S, El-Sabban M, Doughan S, Abou-Kheir W. Molecular mechanisms targeting drug-resistance and metastasis in colorectal cancer: Updates and beyond. World J Gastroenterol 2023; 29:1395-1426. [PMID: 36998426 PMCID: PMC10044855 DOI: 10.3748/wjg.v29.i9.1395] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/12/2022] [Accepted: 11/16/2022] [Indexed: 03/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most diagnosed malignancy and a major leading cause of cancer-related deaths worldwide. Despite advances in therapeutic regimens, the number of patients presenting with metastatic CRC (mCRC) is increasing due to resistance to therapy, conferred by a small population of cancer cells, known as cancer stem cells. Targeted therapies have been highly successful in prolonging the overall survival of patients with mCRC. Agents are being developed to target key molecules involved in drug-resistance and metastasis of CRC, and these include vascular endothelial growth factor, epidermal growth factor receptor, human epidermal growth factor receptor-2, mitogen-activated extracellular signal-regulated kinase, in addition to immune checkpoints. Currently, there are several ongoing clinical trials of newly developed targeted agents, which have shown considerable clinical efficacy and have improved the prognosis of patients who do not benefit from conventional chemotherapy. In this review, we highlight recent developments in the use of existing and novel targeted agents against drug-resistant CRC and mCRC. Furthermore, we discuss limitations and challenges associated with targeted therapy and strategies to combat intrinsic and acquired resistance to these therapies, in addition to the importance of implementing better preclinical models and the application of personalized therapy based on predictive biomarkers for treatment selection.
Collapse
Affiliation(s)
- Samar Al Bitar
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Samer Doughan
- Department of Surgery, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| |
Collapse
|
48
|
Atanasova VS, de Jesus Cardona C, Hejret V, Tiefenbacher A, Mair T, Tran L, Pfneissl J, Draganić K, Binder C, Kabiljo J, Clement J, Woeran K, Neudert B, Wohlhaupter S, Haase A, Domazet S, Hengstschläger M, Mitterhauser M, Müllauer L, Tichý B, Bergmann M, Schweikert G, Hartl M, Dolznig H, Egger G. Mimicking Tumor Cell Heterogeneity of Colorectal Cancer in a Patient-derived Organoid-Fibroblast Model. Cell Mol Gastroenterol Hepatol 2023; 15:1391-1419. [PMID: 36868311 PMCID: PMC10141529 DOI: 10.1016/j.jcmgh.2023.02.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND & AIMS Patient-derived organoid cancer models are generated from epithelial tumor cells and reflect tumor characteristics. However, they lack the complexity of the tumor microenvironment, which is a key driver of tumorigenesis and therapy response. Here, we developed a colorectal cancer organoid model that incorporates matched epithelial cells and stromal fibroblasts. METHODS Primary fibroblasts and tumor cells were isolated from colorectal cancer specimens. Fibroblasts were characterized for their proteome, secretome, and gene expression signatures. Fibroblast/organoid co-cultures were analyzed by immunohistochemistry and compared with their tissue of origin, as well as on gene expression levels compared with standard organoid models. Bioinformatics deconvolution was used to calculate cellular proportions of cell subsets in organoids based on single-cell RNA sequencing data. RESULTS Normal primary fibroblasts, isolated from tumor adjacent tissue, and cancer associated fibroblasts retained their molecular characteristics in vitro, including higher motility of cancer associated compared with normal fibroblasts. Importantly, both cancer-associated fibroblasts and normal fibroblasts supported cancer cell proliferation in 3D co-cultures, without the addition of classical niche factors. Organoids grown together with fibroblasts displayed a larger cellular heterogeneity of tumor cells compared with mono-cultures and closely resembled the in vivo tumor morphology. Additionally, we observed a mutual crosstalk between tumor cells and fibroblasts in the co-cultures. This was manifested by considerably deregulated pathways such as cell-cell communication and extracellular matrix remodeling in the organoids. Thrombospondin-1 was identified as a critical factor for fibroblast invasiveness. CONCLUSION We developed a physiological tumor/stroma model, which will be vital as a personalized tumor model to study disease mechanisms and therapy response in colorectal cancer.
Collapse
Affiliation(s)
- Velina S Atanasova
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | | | - Václav Hejret
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Andreas Tiefenbacher
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Theresia Mair
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Janette Pfneissl
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Kristina Draganić
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Carina Binder
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Julijan Kabiljo
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Clinic of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Janik Clement
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Katharina Woeran
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Barbara Neudert
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | | | - Astrid Haase
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Sandra Domazet
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | | | | | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Boris Tichý
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Michael Bergmann
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Clinic of General Surgery, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Gabriele Schweikert
- Max Planck Institute for Intelligent Systems, Tübingen, Germany; Division of Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Markus Hartl
- Department of Biochemistry and Cell Biology, Max Perutz Labs, Vienna BioCenter (VBC), University of Vienna, Vienna, Austria; Mass Spectrometry Facility, Max Perutz Labs, Vienna BioCenter, University of Vienna, Vienna, Austria
| | - Helmut Dolznig
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria.
| | - Gerda Egger
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Department of Pathology, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
49
|
Zhang J, Zhou N, Deng H, Chen X, Chen Q, Wang Q, Sun L, Wen Y, Cao X, Luo Z, Zhang J, Zhu W, Guo L. Prognostic value of circulating tumor DNA using target next-generation sequencing in extensive-stage small-cell lung cancer. Lung Cancer 2023; 178:11-19. [PMID: 36758321 DOI: 10.1016/j.lungcan.2023.01.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND Chemotherapy remains the mainstay of treatment for small-cell lung cancer (SCLC). Liquid biopsies provide a convenient and non-invasive detection method for monitoring disease progression in patients with SCLC. METHODS We performed next-generation sequencing of 159 plasma samples from 69 patients with extensive-stage (ES)-SCLC. Circulating tumor (ct)DNA levels were quantified in haploid genome equivalents per mL (hGE/mL). MuTect2 was used to detect single nucleotide variants and short insertions/deletions. The "enrichKEGG" function in the "clusterProfiler" R package was used to enrich the mutated genes that only appeared during disease progression. RESULTS In our cohort, 66 of 69 (95.7%) plasma samples at the time of diagnosis had detectable somatic mutations; TP53 (89%) and RB1(56%) were the most frequent mutations, as well as copy number variations in some common SCLC-related genes such as RB1. Combination ctDNA and tissue testing improved the overall detection rate of actionable mutations from 19.4% to 26.9% compared with that of tissue detection alone. In addition, ctDNA levels changed dynamically during the course of treatment and were significantly associated with decreased progression-free survival. Notably, actionable mutations were detected at the time of diagnosis and during disease progression. CONCLUSIONS Our study revealed a dynamic somatic mutation profile through continuous ctDNA detection and confirmed that ctDNA levels can reflect tumor burden and predict PFS in patients with extensive stage-SCLC. Furthermore, we demonstrated that plasma ctDNA assays can provide real-time information on somatic mutations for potential targeted therapies for SCLC.
Collapse
Affiliation(s)
- Jiexia Zhang
- Guangzhou Institute of Respiratory Disease, State Key Laboratory of Respiratory Disease, Department of Respiration, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ningning Zhou
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Huojin Deng
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qunqing Chen
- Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qiongyao Wang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Sun
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Wen
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaolong Cao
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiqiang Luo
- Department of Thoracic Surgery, Maoming People's Hospital, Maoming, Guangdong, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weiliang Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linlang Guo
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
50
|
R HC, Kumar S U, R G, Naayanan PJ, Sathiyarajeswaren P, Devi MSS, K SS, Doss C GP. An integrated investigation of structural and pathway alteration caused by PIK3CA and TP53 mutations identified in cfDNA of metastatic breast cancer. J Cell Biochem 2023; 124:188-204. [PMID: 36563059 DOI: 10.1002/jcb.30354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/12/2022] [Accepted: 11/24/2022] [Indexed: 12/24/2022]
Abstract
In peripheral blood, cell-free DNA (cfDNA) contains circulating tumor DNA (ctDNA), which indicates molecular abnormalities in metastatic breast tumor tissue. The sequencing of cfDNA of Metastatic Breast Cancer (MBC) patients allows assessment of therapy response and noninvasive treatment. In the proposed study, clinically significant alterations in PIK3CA and TP53 genes associated with MBC resulting in a missense substitution of His1047Arg and Arg282Trp from an next-generation sequencing-based multi-gene panel were reported in a cfDNA of a patient with MBC. To investigate the impact of the reported mutation, we used molecular docking, molecular dynamics simulation, network analysis, and pathway analysis. Molecular Docking analysis determined the distinct binding pattern revealing H1047R-ATP complex has a higher number of Hydrogen bonds (H-bonds) and binding affinity with a slight difference compared to the PIK3CA-ATP complex. Following, molecular dynamics simulation for 200 ns, of which H1047R-ATP complex resulted in the instability of PIK3CA. Similarly, for TP53 mutant R282W, the zinc-free state (apo) and zinc-bounded (holo) complexes were investigated for conformational change between apo and holo complexes, of which the holo complex mutant R282W was unstable. To validate the conformational change of PIK3CA and TP53, 80% mutation of H1047R in the kinase domain of p110α expressed ubiquitously in PIK3CA protein that alters PI3K pathway, while R282W mutation in DNA binding helix (H2) region of P53 protein inhibits the transcription factor in P53 pathway causing MBC. According to our findings, the extrinsic (hypoxia, oxidative stress, and acidosis); intrinsic factors (MYC amplification) in PIK3CA and TP53 mutations will provide potential insights for developing novel therapeutic methods for MBC therapy.
Collapse
Affiliation(s)
- Hephzibah Cathryn R
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Udhaya Kumar S
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Gnanasambandan R
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | | | | | - M S Shree Devi
- Siddha Central Research Institute (CCRS), Chennai, Tamil Nadu, India
| | - Satish Srinivas K
- Department of Radiation Oncology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, India
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|