1
|
van Kessel E, Woerdman N, Pluim D, Kerklaan BM, van den Broek D, Huitema ADR, Brandsma D. Added prognostic value of circulating tumor cell numbers in CSF of patients with leptomeningeal metastasis from epithelial tumors. Eur J Cancer 2025; 220:115377. [PMID: 40184847 DOI: 10.1016/j.ejca.2025.115377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Leptomeningeal metastases (LM) from solid tumors are described in up to 10 % of patients, and median survival rates are low. Advanced techniques for circulating tumor cell enumeration in cerebrospinal fluid (CSF) can help in diagnosing patients with LM but also could have value in prognostication. Our aim was to investigate whether the number of circulating tumor cells (CTCs) in CSF are of added value in survival prediction in LM patients. METHODS We performed a single-center retrospective study in a cohort of consecutive patients with LM from epithelial tumors. Circulating tumor cells in CSF were measured by an Epithelial Cell Adhesion Molecule (EpCAM)-based immunoflow cytometry method at LM diagnosing. The added prognostic value of the number of CTCs in CSF on top of known clinical prognostic factors for LM was assessed. We compared Cox proportional hazards regression models with and without CTCs by discriminative performance measures. RESULTS We included 103 eligible patients with epithelial tumors (60 % NSCLC, 29 % breast cancer), who were diagnosed with either possible, probable, or confirmed LM according to EANO-ESMO criteria. Median survival was 129 days. CTC numbers in CSF were of significant added prognostic value in a multivariable prognostic model (HR = 1.86, 95 % CI = 1.26-2.76, likelihood ratio test p-value=0.003 for models with CTCs versus without CTCs). CONCLUSION Our findings indicate that the number of CTCs in CSF, as measured by EpCAM immunoflow cytometry, are of additional prognostic value to other well-known clinical predictors of survival in LM. The number of CTCs in CSF should be taken in account when assessing the prognosis of patients with LM.
Collapse
Affiliation(s)
- Emma van Kessel
- Department of Neurology & Neurosurgery, University Medical Center Utrecht/UMC Utrecht Brain Center, Utrecht University, the Netherlands; Department of Neuro-Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands.
| | - Noor Woerdman
- Department of Neuro-Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Dick Pluim
- Department of Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Bojana Milojkovic Kerklaan
- Department of Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Department of General Practice, Intellectual Disability Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Daan van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Alwin D R Huitema
- Department of Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Dieta Brandsma
- Department of Neuro-Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Juan YC, Chen X, Tseng JY, Lin HJ, Hung CH, Hsueh PR, Lin JJ, Cho DY, Chen CC. Beyond the blood-brain barrier: feasibility and technical validation of dual-compartment circulating tumor cells detection in high-grade glioma patients. Neurosurg Rev 2025; 48:359. [PMID: 40214852 PMCID: PMC11991960 DOI: 10.1007/s10143-025-03511-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/18/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025]
Abstract
The elusive nature of brain tumor progression, hidden behind the blood-brain barrier, presents significant challenges for treatment monitoring in high-grade gliomas. In this feasibility study, we evaluate a novel approach to tracking glioblastoma through liquid biopsy, assessing whether tumor cells leave detectable molecular footprints in both blood and cerebrospinal fluid (CSF). Using the MiSelect R II System with specialized microfluidic technology, we analyzed paired blood and CSF samples from six glioblastoma patients, revealing a striking presence of circulating tumor cells (CTCs)- with higher abundance in CSF, where detection rates reached 100% compared to 83.3% in blood. Our technical validation demonstrates the system's capability to identify CTCs through multi-marker analysis (EGFR+/GFAP+/CD45-). Preliminary observations revealed higher CTC counts in CSF (median 15.5 cells/mL) compared to blood (median 3.0 cells/mL), with notable differences between compartments suggesting they may reflect distinct aspects of disease biology. In a patient who developed progressive disease, we observed a substantial increase in CSF CTCs from 14 to 116 cells/mL, warranting further investigation in larger cohorts. Additionally, we detected CTC clusters in both compartments, an intriguing finding with potential biological significance. While our interim analysis provides technical proof-of-concept for CTC detection in glioblastoma patients, the limited sample size precludes definitive conclusions regarding clinical utility. These findings establish a methodological foundation for future comprehensive studies exploring the relationship between CTC dynamics and clinical outcomes in high-grade gliomas.
Collapse
Affiliation(s)
- Yu-Chung Juan
- Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan
- Neuroscience and Brain Disease Center, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - XianXiu Chen
- Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan
- Neuroscience and Brain Disease Center, China Medical University, Taichung, Taiwan
| | | | - Hui-Ju Lin
- Department of Laboratory Medicine, China Medical University and Hospital, Taichung, Taiwan
| | - Cheng-Hao Hung
- Department of Laboratory Medicine, China Medical University and Hospital, Taichung, Taiwan
| | - Po-Ren Hsueh
- Department of Laboratory Medicine, China Medical University and Hospital, Taichung, Taiwan
- Department of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Jung-Ju Lin
- Sleep Medicine Center, China Medical University Hospital, Taichung, Taiwan
| | - Der-Yang Cho
- Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan
| | - Chun-Chung Chen
- Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan.
- Neuroscience and Brain Disease Center, China Medical University, Taichung, Taiwan.
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan.
| |
Collapse
|
3
|
Sener U, Wilcox JA, Boire AA. Leptomeningeal Disease: Current Approaches and Future Directions. Curr Neurol Neurosci Rep 2025; 25:25. [PMID: 40100294 PMCID: PMC11920312 DOI: 10.1007/s11910-025-01412-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE OF REVIEW Leptomeningeal disease (LMD), or spread of cancer cells into the pia and arachnoid membranes encasing the brain and spinal cord, is associated with high symptom burden and poor survival at 2 to 5 months. Conventional treatments including photon-based radiation therapy, systemic chemotherapy, and intrathecal chemotherapy demonstrate limited efficacy. Despite significant successes for a range of solid tumors, immunotherapy has not yet demonstrated significant efficacy in management of LMD. Advances in understanding of LMD pathophysiology, improved diagnostics, and novel therapeutics are shifting this paradigm. In this article, we review diagnostic and treatment challenges associated with LMD. RECENT FINDINGS We discuss the use of novel cerebrospinal fluid (CSF) analysis techniques such as circulating tumor cell and CSF cell-free DNA assessment to overcome limitations of conventional diagnostic modalities. We then review advances in treatment including clinical trial data demonstrating efficacy of proton craniospinal radiation to treat the entire neuroaxis. We discuss emerging data regarding targeted therapeutics conferring durable survival benefit. Novel therapeutics and combinatorial treatment approaches will likely further improve outcomes for patients with LMD.
Collapse
Affiliation(s)
- Ugur Sener
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Jessica A Wilcox
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adrienne A Boire
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Brain Tumor Center, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
4
|
Jirattikanwong N, Charoentum C, Phenphinan N, Pooriwarangkakul P, Ruttanaphol D, Phinyo P. Individual survival prediction model for patients with leptomeningeal metastasis. Jpn J Clin Oncol 2025; 55:237-245. [PMID: 39563491 DOI: 10.1093/jjco/hyae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/29/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Survival prediction for patients with leptomeningeal metastasis (LM) is crucial for making proper management plans and counseling patients. Prognostic models in this patient domain have been limited, and existing models often include predictors that are not available in resource-limited settings. Our aim was to develop a practical, individualized survival prediction model for patients diagnosed with LM. METHODS We collected a retrospective cohort of patients diagnosed with LM from cerebrospinal fluid at Chiang Mai University Hospital from January 2015 to July 2021. Nine candidate predictors included male gender, age > 60 years, presence of extracranial involvement, types of primary cancer, the time between primary cancer and LM diagnosis, presence of cerebral symptoms, cranial symptoms, spinal symptoms, and abnormal CSF profiles. Flexible parametric survival analysis was used to develop the survival prognostic model for predicting survival at 3, 6, and 12 months after diagnosis. The model was evaluated for discrimination and calibration. RESULTS 161 patients with 133 events were included. The derived individual survival prediction model for patients with LM, or the LMsurv model, consists of three final predictors: types of primary cancer, presence of cerebral symptoms, and presence of spinal symptoms. The model showed acceptable discrimination (Harrell's C-statistics: 0.72; 95% confidence interval 0.68-0.76) and was well calibrated at 3, 6, and 12 months. CONCLUSIONS The LMsurv model, incorporating three practical predictors, demonstrated acceptable discrimination and calibration for predicting survival in LM patients. This model could serve as an assisting tool during clinical decision-making. External validation is suggested to confirm the generalizability of the model.
Collapse
Affiliation(s)
- Noraworn Jirattikanwong
- Center for Clinical Epidemiology and Clinical Statistics, Faculty of Medicine, Chiang Mai University, 110 Intawaroros, Si Phum, Muang, Chiang Mai 50200, Thailand
- Department of Biomedical Informatics and Clinical Epidemiology (BioCE), Faculty of Medicine, Chiang Mai University, 110 Intawaroros, Si Phum, Muang, Chiang Mai 50200, Thailand
| | - Chaiyut Charoentum
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, 110 Intawaroros, Si Phum, Muang, Chiang Mai 50200, Thailand
| | - Niphitphon Phenphinan
- Department of Orthopedics, Faculty of Medicine, Chiang Mai University, 110 Intawaroros, Si Phum, Muang, Chiang Mai 50200, Thailand
| | - Phurich Pooriwarangkakul
- Department of Psychiatry, Faculty of Medicine, Chiang Mai University, 110 Intawaroros, Si Phum, Muang, Chiang Mai 50200, Thailand
| | - Danusorn Ruttanaphol
- Department of Otolaryngology, Faculty of Medicine, Chiang Mai University, 110 Intawaroros, Si Phum, Muang, Chiang Mai 50200, Thailand
| | - Phichayut Phinyo
- Center for Clinical Epidemiology and Clinical Statistics, Faculty of Medicine, Chiang Mai University, 110 Intawaroros, Si Phum, Muang, Chiang Mai 50200, Thailand
- Department of Biomedical Informatics and Clinical Epidemiology (BioCE), Faculty of Medicine, Chiang Mai University, 110 Intawaroros, Si Phum, Muang, Chiang Mai 50200, Thailand
- Center of Multidisciplinary Technology for Advanced Medicine, Faculty of Medicine, Chiang Mai University, 110 Intawaroros, Si Phum, Muang, Chiang Mai 50200, Thailand
| |
Collapse
|
5
|
Ozair A, Wilding H, Bhanja D, Mikolajewicz N, Glantz M, Grossman SA, Sahgal A, Le Rhun E, Weller M, Weiss T, Batchelor TT, Wen PY, Haas-Kogan DA, Khasraw M, Rudà R, Soffietti R, Vollmuth P, Subbiah V, Bettegowda C, Pham LC, Woodworth GF, Ahluwalia MS, Mansouri A. Leptomeningeal metastatic disease: new frontiers and future directions. Nat Rev Clin Oncol 2025; 22:134-154. [PMID: 39653782 DOI: 10.1038/s41571-024-00970-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2024] [Indexed: 12/12/2024]
Abstract
Leptomeningeal metastatic disease (LMD), encompassing entities of 'meningeal carcinomatosis', neoplastic meningitis' and 'leukaemic/lymphomatous meningitis', arises secondary to the metastatic dissemination of cancer cells from extracranial and certain intracranial malignancies into the leptomeninges and cerebrospinal fluid. The clinical burden of LMD has been increasing secondary to more sensitive diagnostics, aggressive local therapies for discrete brain metastases, and improved management of extracranial disease with targeted and immunotherapeutic agents, resulting in improved survival. However, owing to drug delivery challenges and the unique microenvironment of LMD, novel therapies against systemic disease have not yet translated into improved outcomes for these patients. Underdiagnosis and misdiagnosis are common, response assessment remains challenging, and the prognosis associated with this disease of whole neuroaxis remains extremely poor. The dearth of effective therapies is further challenged by the difficulties in studying this dynamic disease state. In this Review, a multidisciplinary group of experts describe the emerging evidence and areas of active investigation in LMD and provide directed recommendations for future research. Drawing upon paradigm-changing advances in mechanistic science, computational approaches, and trial design, the authors discuss domain-specific and cross-disciplinary strategies for optimizing the clinical and translational research landscape for LMD. Advances in diagnostics, multi-agent intrathecal therapies, cell-based therapies, immunotherapies, proton craniospinal irradiation and ongoing clinical trials offer hope for improving outcomes for patients with LMD.
Collapse
Affiliation(s)
- Ahmad Ozair
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hannah Wilding
- Penn State College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Debarati Bhanja
- Department of Neurosurgery, NYU Langone Medical Center, New York, NY, USA
| | - Nicholas Mikolajewicz
- Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael Glantz
- Department of Neurosurgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Stuart A Grossman
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arjun Sahgal
- Department of Radiation Oncology, Sunnybrook Odette Cancer Center, University of Toronto, Toronto, Ontario, Canada
- Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Emilie Le Rhun
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Tracy T Batchelor
- Center for Neuro-Oncology, Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Daphne A Haas-Kogan
- Center for Neuro-Oncology, Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Mustafa Khasraw
- Preston Robert Tisch Brain Tumour Center at Duke, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science Hospital, Turin, Italy
| | - Riccardo Soffietti
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science Hospital, Turin, Italy
- Department of Oncology, Candiolo Institute for Cancer Research, FPO-IRCCS, Candiolo, Turin, Italy
| | - Philipp Vollmuth
- Division for Computational Radiology and Clinical AI, University Hospital Bonn, Bonn, Germany
- Division for Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Vivek Subbiah
- Early Phase Drug Development Program, Sarah Cannon Research Institute, Nashville, TN, USA
| | - Chetan Bettegowda
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lily C Pham
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
- Brain Tumor Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Brain Tumor Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Manmeet S Ahluwalia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA.
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA.
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
6
|
Bortolot M, Huijs JWJ, Brandsma D, Compter A, van Geel RMJM, Hendriks LEL. Advancing leptomeningeal metastases treatment in EGFR-mutated non-small cell lung cancer: lessons from the BLOSSOM trial. Transl Lung Cancer Res 2025; 14:7-13. [PMID: 39958217 PMCID: PMC11826264 DOI: 10.21037/tlcr-24-1006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/19/2024] [Indexed: 02/18/2025]
Affiliation(s)
- Martina Bortolot
- Department of Pulmonary Diseases, GROW – Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Jarno W. J. Huijs
- Department of Pulmonary Diseases, GROW – Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Dieta Brandsma
- Department of Neuro-Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Annette Compter
- Department of Neuro-Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Robin M. J. M. van Geel
- Department of Clinical Pharmacy & Toxicology, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Lizza E. L. Hendriks
- Department of Pulmonary Diseases, GROW – Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
7
|
Cohen-Nowak AJ, Hill VB, Kumthekar P. Diagnostics and Screening in Breast Cancer with Brain and Leptomeningeal Metastasis: A Review of the Literature. Cancers (Basel) 2024; 16:3686. [PMID: 39518124 PMCID: PMC11545036 DOI: 10.3390/cancers16213686] [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: 08/30/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Brain and leptomeningeal metastases are complications of breast cancer with high rates of morbidity and mortality and have an estimated incidence of up to 30%. While National Comprehensive Cancer Network (NCCN) guidelines recommend screening for central nervous system metastasis in other neurotropic cancers such as non-small cell lung cancer, there are no such recommendations for asymptomatic breast cancer patients at any stage of disease. This review highlights ongoing studies into screening and diagnostics for breast cancer with brain and leptomeningeal metastasis (BCBLM) as they relate to patient outcomes and prognostication. These include imaging methods such as MRI with novel contrast agents with or without PET/CT, as well as 'liquid biopsy' testing of the cerebrospinal fluid and serum to analyze circulating tumor cells, genomic material, proteins, and metabolites. Given recent advances in radiation, neurosurgery, and systemic treatments for BCBLM, screening for CNS involvement should be considered in patients with advanced breast cancer as it may impact treatment decisions and overall survival.
Collapse
Affiliation(s)
- Adam J. Cohen-Nowak
- Department of Internal Medicine, McGaw Medical Center of Northwestern University, Chicago, IL 60611, USA;
| | - Virginia B. Hill
- Neuroradiology Section, Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Priya Kumthekar
- Department of Neurology, Northwestern Memorial Hospital, Chicago, IL 60611, USA
| |
Collapse
|
8
|
Wilcox JA, Chukwueke UN, Ahn MJ, Aizer AA, Bale TA, Brandsma D, Brastianos PK, Chang S, Daras M, Forsyth P, Garzia L, Glantz M, Oliva ICG, Kumthekar P, Le Rhun E, Nagpal S, O'Brien B, Pentsova E, Lee EQ, Remsik J, Rudà R, Smalley I, Taylor MD, Weller M, Wefel J, Yang JT, Young RJ, Wen PY, Boire AA. Leptomeningeal metastases from solid tumors: A Society for Neuro-Oncology and American Society of Clinical Oncology consensus review on clinical management and future directions. Neuro Oncol 2024; 26:1781-1804. [PMID: 38902944 PMCID: PMC11449070 DOI: 10.1093/neuonc/noae103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Indexed: 06/22/2024] Open
Abstract
Leptomeningeal metastases (LM) are increasingly becoming recognized as a treatable, yet generally incurable, complication of advanced cancer. As modern cancer therapeutics have prolonged the lives of patients with metastatic cancer, specifically in patients with parenchymal brain metastases, treatment options, and clinical research protocols for patients with LM from solid tumors have similarly evolved to improve survival within specific populations. Recent expansions in clinical investigation, early diagnosis, and drug development have given rise to new unanswered questions. These include leptomeningeal metastasis biology and preferred animal modeling, epidemiology in the modern cancer population, ensuring validation and accessibility of newer leptomeningeal metastasis diagnostics, best clinical practices with multimodality treatment options, clinical trial design and standardization of response assessments, and avenues worthy of further research. An international group of multi-disciplinary experts in the research and management of LM, supported by the Society for Neuro-Oncology and American Society of Clinical Oncology, were assembled to reach a consensus opinion on these pressing topics and provide a roadmap for future directions. Our hope is that these recommendations will accelerate collaboration and progress in the field of LM and serve as a platform for further discussion and patient advocacy.
Collapse
Affiliation(s)
- Jessica A Wilcox
- Department of Neurology, Brain Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ugonma N Chukwueke
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ayal A Aizer
- Department of Radiation Oncology, Brigham and Women's Hospital / Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Tejus A Bale
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Dieta Brandsma
- Department of Neuro-Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Priscilla K Brastianos
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Chang
- Division of Neuro-Oncology, Department of Neurosurgery, University of San Francisco California, San Francisco, California, USA
| | - Mariza Daras
- Department of Neurology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Peter Forsyth
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Livia Garzia
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael Glantz
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Isabella C Glitza Oliva
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Priya Kumthekar
- The Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Emilie Le Rhun
- Departments of Neurology and Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Seema Nagpal
- Division of Neuro-Oncology, Department of Neurology, Stanford University School of Medicine, Stanford, California, USA
| | - Barbara O'Brien
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elena Pentsova
- Department of Neurology, Brain Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Eudocia Quant Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jan Remsik
- Laboratory for Immunology of Metastatic Ecosystems, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience, University and City of Health and Science Hospital, Turin, Italy
- Department of Neurology, Castelfranco Veneto and Treviso Hospitals, Castelfranco Veneto, Italy
| | - Inna Smalley
- Department of Tumor Biology, The Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Michael D Taylor
- Division of Neurosurgery, Department of Surgery, Texas Children's Hospital, Houston, Texas, USA
- Neuro-oncology Research Program, Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Jeffrey Wefel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jonathan T Yang
- Department of Radiation Oncology, Department of Radiation Oncology, New York University School of Medicine, New York, New York, USA
| | - Robert J Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Adrienne A Boire
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Neurology, Brain Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
9
|
Jeng MY, Yu HA. Leptomeningeal Metastasis in the Era of CNS-Penetrant Tyrosine Kinase Inhibitor Therapy. J Clin Oncol 2024; 42:2727-2730. [PMID: 38941569 DOI: 10.1200/jco.24.01061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 06/30/2024] Open
Affiliation(s)
- Mark Y Jeng
- Department of Medicine, Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Helena A Yu
- Department of Medicine, Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
10
|
Kumthekar PU, Blouw B, Corkos P, Nagpal S, Tripathy A, Piccioni D, Youssef M. The HER2 flip-HER2 amplification of tumor cells in the cerebrospinal fluid of breast cancer patients with leptomeningeal disease: implications for treating the LM tumor with anti-HER2 therapy. Front Oncol 2024; 14:1402651. [PMID: 38826788 PMCID: PMC11140729 DOI: 10.3389/fonc.2024.1402651] [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: 03/18/2024] [Accepted: 04/15/2024] [Indexed: 06/04/2024] Open
Abstract
Introduction CNSide is a platform that detects and characterizes tumor cells in the cerebrospinal fluid (CSF) of patients with leptomeningeal disease (LMD). The platform was validated per College of American Pathologists (CAP) and Clinical Laboratories Improvement Amendment (CLIA) guidelines and run as a commercial Laboratory Developed Test (LDT) at Biocept in San Diego, CA. The platform allows CSF tumor cell (CSF-TC) enumeration and biomarker characterization by fluorescent in situ hybridization (FISH). Methods We performed a multicenter retrospective chart review of HER2 FISH CNSide test results that were commercially ordered on 26 patients by physicians for LMD breast cancer patients between April 2020 and October 2022. Results We show that HER2 is amplified on CSF tumor cells in 62% (16/26) of LMD breast cancer patients. 10/26 (38%) patients had discordant HER2-positivity between the primary tumor tissue and CSF-TC; of these, 35% (9/26) of the patients displayed HER2 amplification on the CSF-TCs, however were categorized as HER2 negative on the primary tumor. Of the 27% (7/26) patients with a HER2 positive primary tumor, one patient showed a HER2 negative LMD tumor. Two patients, 8% (2/26) had a HER2 equivocal primary tumor; of these, one demonstrated a HER2 negative, and one a HER2 positive LMD tumor. Serial analysis (at least 4 longitudinal tests) of HER2 status of the CSF-TC throughout therapy was available for 14 patients and demonstrated that HER2 status of the LMD changed in 29% (4/14) during their treatment course and impacted care decisions. Conclusions Our data suggests that CSF-TC HER2 FISH analysis in LMD breast cancer patients may be discordant to the primary tumor sample and the discovery of HER2 positivity in the CSF may open doors to anti-HER2 targeted therapy options for LMD patients.
Collapse
Affiliation(s)
- Priya U. Kumthekar
- Neurology (Neuro-Oncology) and Medicine (Hematology and Oncology), Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute, Chicago, IL, United States
| | | | | | - Seema Nagpal
- Neurology and Neurological Sciences, Neurosurgery, Stanford University Medicine, Stanford, CA, United States
| | - Arushi Tripathy
- Neurosurgery, Michigan University Medicine, Ann Arbor, MI, United States
| | - David Piccioni
- Neurosciences, Neuro-Oncology, University of San Diego Health, San Diego, CA, United States
| | - Michael Youssef
- Department of Neurology and Hematology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
11
|
Barbour AB, Blouw B, Taylor LP, Graber JJ, McGranahan T, Blau M, Halasz LM, Lo SS, Tseng YD, Venur V, Yang JT. Prognostic value of cerebrospinal fluid tumor cell count in leptomeningeal disease from solid tumors. J Neurooncol 2024; 167:509-514. [PMID: 38441840 DOI: 10.1007/s11060-024-04615-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 02/19/2024] [Indexed: 05/16/2024]
Abstract
PURPOSE Treatment decisions for leptomeningeal disease (LMD) rely on patient risk stratification, since clinicians lack objective prognostic tools. The introduction of rare cell capture technology for identification of cerebrospinal fluid tumor cells (CSF-TCs), such as CNSide assay, improved the sensitivity of LMD diagnosis, but prognostic value is unknown. This study assesses the prognostic value of CSF-TC density in patients with LMD from solid tumors. METHODS We conducted a retrospective cohort study of patients with newly diagnosed or previously treated LMD from a single institution who had CNSide assay testing for CSF-TCs from 2020 to 2023. Univariable and multivariable survival analyses were conducted with Cox proportional-hazards modeling. Maximally-selected rank statistics were used to determine an optimal cutpoint for CSF-TC density and survival. RESULTS Of 31 patients, 29 had CSF-TCs detected on CNSide. Median (interquartile range [IQR]) CSF-TC density was 67.8 (4.7-639) TCs/mL. CSF cytology was positive in 16 of 29 patients with positive CNSide (CNSide diagnostic sensitivity = 93.5%, negative predictive value = 85.7%). Median (IQR) survival from time of CSF-TC detection was 176 (89-481) days. On univariable and multivariable analysis, CSF-TC density was significantly associated with survival. An optimal cutpoint for dichotomizing survival by CSF-TC density was 19.34 TCs/mL. The time-dependent sensitivity and specificity for survival using this stratification were 76% and 67% at 6 months and 65% and 67% at 1 year, respectively. CONCLUSIONS CSF-TC density may carry prognostic value in patients with LMD from solid tumors. Integrating CSF-TC density into LMD patient risk-stratification may help guide treatment decisions.
Collapse
Affiliation(s)
- Andrew B Barbour
- Department of Radiation Oncology, University of Washington- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Lynne P Taylor
- Department of Neurology, University of Washington- Alvord Brain Tumor Center, Seattle, WA, USA
| | - Jerome J Graber
- Department of Neurology, University of Washington- Alvord Brain Tumor Center, Seattle, WA, USA
| | - Tresa McGranahan
- Division of Hematology-Oncology, Scripps Cancer Center, La Jolla, CA, USA
| | - Molly Blau
- Department of Radiation Oncology, University of Washington- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lia M Halasz
- Department of Radiation Oncology, University of Washington- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Simon S Lo
- Department of Radiation Oncology, University of Washington- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yolanda D Tseng
- Department of Radiation Oncology, University of Washington- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Vyshak Venur
- Department of Neurology, University of Washington- Alvord Brain Tumor Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jonathan T Yang
- Department of Radiation Oncology, University of Washington- Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
12
|
Diaz M, Chudsky S, Pentsova E, Miller AM. Clinical applications of cerebrospinal fluid liquid biopsies in central nervous system tumors. Transl Oncol 2024; 41:101881. [PMID: 38218027 PMCID: PMC10825768 DOI: 10.1016/j.tranon.2024.101881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/28/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024] Open
Abstract
For patients with central nervous system (CNS) malignancies, liquid biopsies of the cerebrospinal fluid (CSF) may offer an unparalleled source of information about the tumor, with much less risk than traditional biopsies. Two techniques have been adapted to CSF in clinical settings: circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). CTCs have been employed mostly as a diagnostic tool for leptomeningeal metastases in epithelial tumors, although they may also have value in the prognostication and monitoring of this disease. The ctDNA technology has been studied in a variety of primary and metastatic brain and spinal cord tumors, where it can be used for diagnosis and molecular classification, with some work suggesting that it may also be useful for longitudinal tracking of tumor evolution or as a marker of residual disease. This review summarizes recent publications on the use of these two tests in CSF, focusing on their established and potential clinical applications.
Collapse
Affiliation(s)
- Maria Diaz
- Department of Neurology, Division of Neuro-Oncology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sofia Chudsky
- Office of Professional Development, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hunter College, New York, NY, USA
| | - Elena Pentsova
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexandra M Miller
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
13
|
Barbour AB, Kotecha R, Lazarev S, Palmer JD, Robinson T, Yerramilli D, Yang JT. Radiation Therapy in the Management of Leptomeningeal Disease From Solid Tumors. Adv Radiat Oncol 2024; 9:101377. [PMID: 38405313 PMCID: PMC10885590 DOI: 10.1016/j.adro.2023.101377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/03/2023] [Indexed: 02/27/2024] Open
Abstract
Purpose Leptomeningeal disease (LMD) is clinically detected in 5% to 10% of patients with solid tumors and is a source of substantial morbidity and mortality. Prognosis for this entity remains poor and treatments are palliative. Radiation therapy (RT) is an essential tool in the management of LMD, and a recent randomized trial demonstrated a survival benefit for proton craniospinal irradiation (CSI) in select patients. In the setting of this recent advance, we conducted a review of the role of RT in LMD from solid tumors to evaluate the evidence basis for RT recommendations. Methods and Materials In November 2022, we conducted a comprehensive literature search in PubMed, as well as a review of ongoing clinical trials listed on ClinicalTrials.gov, to inform a discussion on the role of RT in solid tumor LMD. Because of the paucity of high-quality published evidence, discussion was informed more by expert consensus and opinion, including a review of societal guidelines, than evidence from clinical trials. Results Only 1 prospective randomized trial has evaluated RT for LMD, demonstrating improved central nervous system progression-free survival for patients with breast and lung cancer treated with proton CSI compared with involved-field RT. Modern photon CSI techniques have improved upon historical rates of acute hematologic toxicity, but the overall benefit of this modality has not been prospectively evaluated. Multiple retrospective studies have explored the use of involved-field RT or the combination of RT with chemotherapy, but clear evidence of survival benefit is lacking. Conclusions Optimal management of LMD with RT remains reliant upon expert opinion, with proton CSI indicated in patients with good performance status and extra-central nervous system disease that is either well-controlled or for which effective treatment options are available. Photon-based CSI traditionally has been associated with increased marrow and gastrointestinal toxicities, though intensity modulated RT/volumetric-modulated arc therapy based photon CSI may have reduced the toxicity profile. Further work is needed to understand the role of radioisotopes as well as combined modality treatment with intrathecal or central nervous system penetrating systemic therapies.
Collapse
Affiliation(s)
- Andrew B. Barbour
- Department of Radiation Oncology, University of Washington – Fred Hutchinson Cancer Center, Seattle, Washington
| | - Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida
| | - Stanislav Lazarev
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joshua D. Palmer
- Department of Radiation Oncology, The James Cancer Hospital, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Timothy Robinson
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut
| | - Divya Yerramilli
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jonathan T. Yang
- Department of Radiation Oncology, University of Washington – Fred Hutchinson Cancer Center, Seattle, Washington
| |
Collapse
|
14
|
Goldberg M, Mondragon-Soto MG, Altawalbeh G, Meyer B, Aftahy AK. New Breakthroughs in the Diagnosis of Leptomeningeal Carcinomatosis: A Review of Liquid Biopsies of Cerebrospinal Fluid. Cureus 2024; 16:e55187. [PMID: 38558729 PMCID: PMC10980855 DOI: 10.7759/cureus.55187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Leptomeningeal carcinomatosis represents a terminal stage and is a devastating complication of cancer. Despite its high incidence, current diagnostic methods fail to accurately detect this condition in a timely manner. This failure to diagnose leads to the refusal of treatment and the absence of clinical trials, hampering the development of new therapy strategies. The use of liquid biopsy is revolutionizing the field of diagnostic oncology. The dynamic and non-invasive detection of tumor markers has enormous potential in cancer diagnostics and treatment. Leptomeningeal carcinomatosis is a condition where invasive tissue biopsy is not part of the routine diagnostic analysis, making liquid biopsy an essential diagnostic tool. Several elements in cerebrospinal fluid (CSF) have been investigated as potential targets of liquid biopsy, including free circulating tumor cells, free circulating nucleic acids, proteins, exosomes, and even non-tumor cells as part of the dynamic tumor microenvironment. This review aims to summarize current breakthroughs in the research on liquid biopsy, including the latest breakthroughs in the identification of tumor cells and nucleic acids, and give an overview of future directions in the diagnosis of leptomeningeal carcinomatosis.
Collapse
Affiliation(s)
- Maria Goldberg
- Department of Neurosurgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, DEU
| | | | - Ghaith Altawalbeh
- Department of Neurosurgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, DEU
| | - Bernhard Meyer
- Department of Neurosurgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, DEU
| | - Amir Kaywan Aftahy
- Department of Neurosurgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, DEU
| |
Collapse
|
15
|
Pellerino A, Bertero L, Pronello E, Rudà R, Soffietti R. The early recognition and diagnosis of neoplastic meningitis. Expert Rev Neurother 2024; 24:105-116. [PMID: 38145502 DOI: 10.1080/14737175.2023.2295999] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/06/2023] [Indexed: 12/27/2023]
Abstract
INTRODUCTION The diagnosis and monitoring of leptomeningeal metastases (LM) from solid tumors are challenging, and the combination of neurological symptoms, MRI findings, and cerebrospinal fluid (CSF) cytology does not always allow to achieve a definitive diagnosis. AREAS COVERED This review summarizes the studies that have investigated CSF liquid biopsy to improve the initial diagnosis of LM in case the CSF cytology is negative or only suspicious for tumor cells, and monitoring of tumor response following targeted therapies or immunotherapy. In this regard, the early detection of LM recurrence and the development of resistant mutations are critical issues. Moreover, the early identification of subgroups of patients with a higher risk of LM progression, as well as the correlation of LM burden with survival, are discussed. EXPERT OPINION There is an urgent need of prospective studies to monitor longitudinally LM using CSF liquid biopsy and investigate the role of CTC, ctDNA or novel assays. The optimal setting for the longitudinal CSF and blood collection can be clinical trials focused on the molecular diagnosis of LM as well as the response and monitoring following targeted agents.
Collapse
Affiliation(s)
- Alessia Pellerino
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science Hospital, Turin, Italy
| | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Edoardo Pronello
- Neurology Unit, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science Hospital, Turin, Italy
| | - Riccardo Soffietti
- Department of Oncology, Candiolo Institute for Cancer Research, FPO-IRCCS, Turin, Candiolo, Italy
| |
Collapse
|
16
|
Puri S, Malani R, Chalmers A, Kerrigan K, Patel SB, Monynahan K, Cannon L, Blouw B, Akerley W. Keeping a track on leptomeningeal disease in non-small cell lung cancer: A single-institution experience with CNSide TM. Neurooncol Adv 2024; 6:vdad150. [PMID: 38196737 PMCID: PMC10776200 DOI: 10.1093/noajnl/vdad150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024] Open
Abstract
Background Leptomeningeal disease (LMD) is a devastating complication for patients with advanced cancer. Diagnosis and monitoring the response to therapy remains challenging due to limited sensitivity and specificity of standard-of-care (SOC) diagnostic modalities, including cerebrospinal fluid (CSF) cytology, MRI, and clinical evaluation. These hindrances contribute to the poor survival of LMD patients. CNSide is a CLIA-validated test that detects and characterizes CSF-derived tumor cells and cell-free (cf) DNA. We performed a retrospective analysis on the utility of CNSide to analyze CSF obtained from advanced non-small cell lung cancer (aNSCLC) patients with suspected LMD treated at the Huntsman Cancer Institute in Salt Lake City, UT. Methods CNSide was used to evaluate CSF from 15 patients with aNSCLC. CSF tumor cell quantification was performed throughout treatment for 5 patients. CSF tumor cells and cfDNA were characterized for actionable mutations. Results In LMD-positive patients, CNSide detected CSF tumor cells in 88% (22/25) samples versus 40% (10/25) for cytology (matched samples). CSF tumor cell numbers tracked response to therapy in 5 patients where CNSide was used to quantify tumor cells throughout treatment. In 75% (9/12) of the patients, genetic alterations were detected in CSF, with the majority representing gene mutations and amplifications with therapeutic potential. The median survival for LMD patients was 16.1 m (5.2-NR). Conclusions We show that CNSide can supplement the management of LMD in conjunction with SOC methods for the diagnosis, monitoring response to therapy, and identifying actionable mutations unique to the CSF in patients with LMD.
Collapse
Affiliation(s)
- Sonam Puri
- Division of Medical Oncology, The Huntsman Cancer Institute at The University of Utah, Salt Lake City, Utah, USA
| | - Rachna Malani
- Division of Medical Oncology, The Huntsman Cancer Institute at The University of Utah, Salt Lake City, Utah, USA
| | - Anna Chalmers
- Division of Medical Oncology, The Huntsman Cancer Institute at The University of Utah, Salt Lake City, Utah, USA
| | - Kathleen Kerrigan
- Division of Medical Oncology, The Huntsman Cancer Institute at The University of Utah, Salt Lake City, Utah, USA
| | - Shiven B Patel
- Division of Medical Oncology, The Huntsman Cancer Institute at The University of Utah, Salt Lake City, Utah, USA
| | - Kelly Monynahan
- Division of Medical Oncology, The Huntsman Cancer Institute at The University of Utah, Salt Lake City, Utah, USA
| | - Laura Cannon
- Division of Medical Oncology, The Huntsman Cancer Institute at The University of Utah, Salt Lake City, Utah, USA
| | | | - Wallace Akerley
- Division of Medical Oncology, The Huntsman Cancer Institute at The University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
17
|
Malani R, Bhatia A, Warner AB, Yang JT. Leptomeningeal Carcinomatosis from Solid Tumor Malignancies: Treatment Strategies and Biomarkers. Semin Neurol 2023; 43:859-866. [PMID: 37989214 DOI: 10.1055/s-0043-1776996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Leptomeningeal metastases/diseases (LMDs) are a late-stage complication of solid tumor or hematologic malignancies. LMD is spread of cancer cells to the layers of the leptomeninges (pia and arachnoid maters) and subarachnoid space seen in 3 to 5% of cancer patients. It is a disseminated disease which carries with it significant neurologic morbidity and mortality. Our understanding of disease pathophysiology is currently lacking; however, advances are being made. As our knowledge of disease pathogenesis has improved, treatment strategies have evolved. Mainstays of treatment such as radiotherapy have changed from involved-field radiotherapy strategies to proton craniospinal irradiation which has demonstrated promising results in recent clinical trials. Systemic treatment strategies have also improved from more traditional chemotherapeutics with limited central nervous system (CNS) penetration to more targeted therapies with better CNS tumor response. Many challenges remain from earlier clinical detection of disease through improvement of active treatment options, but we are getting closer to meaningful treatment.
Collapse
Affiliation(s)
- Rachna Malani
- Department of Neurosurgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Ankush Bhatia
- Department of Neurology, Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Allison Betof Warner
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | - Jonathan T Yang
- Department of Radiation Oncology, Fred Hutchinson Cancer Center, University of Washington, Seattle, Washington
| |
Collapse
|
18
|
Chew SM, Seidman AD. New strategies for the treatment of breast cancer with leptomeningeal metastasis. Curr Opin Oncol 2023; 35:500-506. [PMID: 37820086 DOI: 10.1097/cco.0000000000000988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
PURPOSE OF REVIEW Leptomeningeal metastasis is a complication of metastatic breast cancer that has a rising incidence likely due to the increased availability of novel systemic therapies, which have improved survival with better extracranial disease control but with limited intracranial efficacy. A poor prognosis of less than 6 months has historically been associated with leptomeningeal metastasis and it is often an exclusion factor for enrollment in clinical trials. There are limited evidence-based data supporting use of therapeutics in leptomeningeal metastasis patients and recommendations are largely derived from retrospective reports and small prospective studies. However, in recent years, there has been a surge in effective modern therapeutics with promising intracranial activity. RECENT FINDINGS The study aims to review the most recent updates in the management of leptomeningeal metastasis in breast cancer. We discuss the effectiveness and limitations of intrathecal administration, predictive biomarkers in the cerebrospinal fluid, proton radiation therapy and promising new systemic therapies such as antibody drug conjugates. SUMMARY Ongoing development of clinical trials that allow inclusion of leptomeningeal metastasis are essential for establishing efficacy potential and discovering new treatment options in this population of great unmet need.
Collapse
Affiliation(s)
- Sonya M Chew
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | |
Collapse
|
19
|
de Jong AC, Isebia KT, Ling SW, de Weerd V, Van NM, Kraan J, Martens JWM, Mehra N, Hamberg P, Lolkema MP, de Wit R, van der Veldt AAM, Wilting SM. Liquid Biopsies for Early Response Evaluation of Radium-223 in Metastatic Prostate Cancer. JCO Precis Oncol 2023; 7:e2300156. [PMID: 38061007 DOI: 10.1200/po.23.00156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/07/2023] [Accepted: 09/26/2023] [Indexed: 12/18/2023] Open
Abstract
PURPOSE Reliable biomarkers for response monitoring during radium-223 treatment in patients with metastatic castration-resistant prostate cancer (mCRPC) are lacking. Circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA), obtained from liquid biopsies, are shown to have prognostic value in mCRPC. The aim of this study was to determine the value of CTCs and ctDNA for response evaluation of radium-223. METHODS In this prospective multicenter study, longitudinal blood draws and imaging were performed in 28 patients with mCRPC and predominantly bone disease, who were treated with radium-223. CTCs were counted (CELLSEARCH CTC test), while fraction of ctDNA was estimated by measuring aneuploidy of cell-free DNA (cfDNA; modified Fast Aneuploidy Screening Test-Sequencing System). CTC counts and aneuploidy score (AS) were categorized as low (<5) and high (≥5). Primary and secondary clinical end points were failure-free survival (FFS), and overall survival (OS) and development of extraosseous metastases, respectively. Additionally, CTC count and AS were related to alkaline phosphatase (ALP) and total tumor volume in bone (TTVbone) on positron emission tomography-computed tomography with 68gallium prostate-specific membrane antigen. RESULTS FFS was longer in patients with a low CTC count or AS either at baseline or after 12 weeks, whereas for OS, only a significant association with CTC count was observed. Liquid biopsy results correlated well with ALP and TTVbone at baseline, but not with change in both parameters after three cycles of radium-223. AS and CTC count were significantly correlated. CONCLUSION CTC count and AS of cfDNA at baseline and during treatment predict clinical response to radium-223 in patients with mCRPC, warranting future evaluation of their value in treatment guidance.
Collapse
Affiliation(s)
- Anouk C de Jong
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Khrystany T Isebia
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Sui Wai Ling
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Vanja de Weerd
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ngoc M Van
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Jaco Kraan
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud UMC, Nijmegen, the Netherlands
| | - Paul Hamberg
- Department of Internal Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, the Netherlands
| | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ronald de Wit
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Saskia M Wilting
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| |
Collapse
|
20
|
Steininger J, Gellrich FF, Engellandt K, Meinhardt M, Westphal D, Beissert S, Meier F, Glitza Oliva IC. Leptomeningeal Metastases in Melanoma Patients: An Update on and Future Perspectives for Diagnosis and Treatment. Int J Mol Sci 2023; 24:11443. [PMID: 37511202 PMCID: PMC10380419 DOI: 10.3390/ijms241411443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Leptomeningeal disease (LMD) is a devastating complication of cancer with a particularly poor prognosis. Among solid tumours, malignant melanoma (MM) has one of the highest rates of metastasis to the leptomeninges, with approximately 10-15% of patients with advanced disease developing LMD. Tumour cells that metastasise to the brain have unique properties that allow them to cross the blood-brain barrier, evade the immune system, and survive in the brain microenvironment. Metastatic colonisation is achieved through dynamic communication between metastatic cells and the tumour microenvironment, resulting in a tumour-permissive milieu. Despite advances in treatment options, the incidence of LMD appears to be increasing and current treatment modalities have a limited impact on survival. This review provides an overview of the biology of LMD, diagnosis and current treatment approaches for MM patients with LMD, and an overview of ongoing clinical trials. Despite the still limited efficacy of current therapies, there is hope that emerging treatments will improve the outcomes for patients with LMD.
Collapse
Affiliation(s)
- Julian Steininger
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität (TU) Dresden, 01307 Dresden, Germany
| | - Frank Friedrich Gellrich
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität (TU) Dresden, 01307 Dresden, Germany
| | - Kay Engellandt
- Department of Neuroradiology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität (TU) Dresden, 01307 Dresden, Germany
| | - Matthias Meinhardt
- Institute of Pathology, University Hospital Carl Gustav Carus, Technische Universität (TU) Dresden, 01307 Dresden, Germany
| | - Dana Westphal
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität (TU) Dresden, 01307 Dresden, Germany
| | - Stefan Beissert
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität (TU) Dresden, 01307 Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität (TU) Dresden, 01307 Dresden, Germany
- Skin Cancer Center at the University Cancer Center, National Center for Tumor Diseases (NCT/UCC), 01307 Dresden, Germany
| | - Isabella C Glitza Oliva
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
21
|
Morganti S, Parsons HA, Lin NU, Grinshpun A. Liquid biopsy for brain metastases and leptomeningeal disease in patients with breast cancer. NPJ Breast Cancer 2023; 9:43. [PMID: 37225714 DOI: 10.1038/s41523-023-00550-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
A significant subset of patients with metastatic breast cancer develops brain metastasis. As efficacy of systemic therapies has improved and patients live longer with metastatic breast cancer, the incidence of breast cancer brain metastases has increased. Brain metastases pose a clinical challenge in diagnosis, treatment, and monitoring across all breast cancer subtypes, and better tools are needed. Liquid biopsy, which enables minimally invasive sampling of a patient's cancer, has the potential to shed light on intra-cranial tumor biology and to improve patient care by enabling therapy tailoring. Here we review current evidence for the clinical validity of liquid biopsy in patients with breast cancer brain metastases, with a focus on circulating tumor cells and circulating tumor DNA.
Collapse
Affiliation(s)
- Stefania Morganti
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Heather A Parsons
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Nancy U Lin
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Albert Grinshpun
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Khaled ML, Tarhini AA, Forsyth PA, Smalley I, Piña Y. Leptomeningeal Disease (LMD) in Patients with Melanoma Metastases. Cancers (Basel) 2023; 15:cancers15061884. [PMID: 36980770 PMCID: PMC10047692 DOI: 10.3390/cancers15061884] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Leptomeningeal disease (LMD) is a devastating complication caused by seeding malignant cells to the cerebrospinal fluid (CSF) and the leptomeningeal membrane. LMD is diagnosed in 5-15% of patients with systemic malignancy. Management of LMD is challenging due to the biological and metabolic tumor microenvironment of LMD being largely unknown. Patients with LMD can present with a wide variety of signs and/or symptoms that could be multifocal and include headache, nausea, vomiting, diplopia, and weakness, among others. The median survival time for patients with LMD is measured in weeks and up to 3-6 months with aggressive management, and death usually occurs due to progressive neurologic dysfunction. In melanoma, LMD is associated with a suppressive immune microenvironment characterized by a high number of apoptotic and exhausted CD4+ T-cells, myeloid-derived suppressor cells, and a low number of CD8+ T-cells. Proteomics analysis revealed enrichment of complement cascade, which may disrupt the blood-CSF barrier. Clinical management of melanoma LMD consists primarily of radiation therapy, BRAF/MEK inhibitors as targeted therapy, and immunotherapy with anti-PD-1, anti-CTLA-4, and anti-LAG-3 immune checkpoint inhibitors. This review summarizes the biology and anatomic features of melanoma LMD, as well as the current therapeutic approaches.
Collapse
Affiliation(s)
- Mariam Lotfy Khaled
- Metabolism and Physiology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo 12613, Egypt
| | - Ahmad A Tarhini
- Departments of Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Peter A Forsyth
- Neuro-Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Inna Smalley
- Metabolism and Physiology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Yolanda Piña
- Neuro-Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
23
|
Yang JT, Wijetunga NA, Pentsova E, Wolden S, Young RJ, Correa D, Zhang Z, Zheng J, Steckler A, Bucwinska W, Bernstein A, Betof Warner A, Yu H, Kris MG, Seidman AD, Wilcox JA, Malani R, Lin A, DeAngelis LM, Lee NY, Powell SN, Boire A. Randomized Phase II Trial of Proton Craniospinal Irradiation Versus Photon Involved-Field Radiotherapy for Patients With Solid Tumor Leptomeningeal Metastasis. J Clin Oncol 2022; 40:3858-3867. [PMID: 35802849 PMCID: PMC9671756 DOI: 10.1200/jco.22.01148] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 05/26/2022] [Accepted: 06/14/2022] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Photon involved-field radiotherapy (IFRT) is the standard-of-care radiotherapy for patients with leptomeningeal metastasis (LM) from solid tumors. We tested whether proton craniospinal irradiation (pCSI) encompassing the entire CNS would result in superior CNS progression-free survival (PFS) compared with IFRT. PATIENTS AND METHODS We conducted a randomized, phase II trial of pCSI versus IFRT in patients with non-small-cell lung cancer and breast cancers with LM. We enrolled patients with other solid tumors to an exploratory pCSI group. For the randomized groups, patients were assigned (2:1), stratified by histology and systemic disease status, to pCSI or IFRT. The primary end point was CNS PFS. Secondary end points included overall survival (OS) and treatment-related adverse events (TAEs). RESULTS Between April 16, 2020, and October 11, 2021, 42 and 21 patients were randomly assigned to pCSI and IFRT, respectively. At planned interim analysis, a significant benefit in CNS PFS was observed with pCSI (median 7.5 months; 95% CI, 6.6 months to not reached) compared with IFRT (2.3 months; 95% CI, 1.2 to 5.8 months; P < .001). We also observed OS benefit with pCSI (9.9 months; 95% CI, 7.5 months to not reached) versus IFRT (6.0 months; 95% CI, 3.9 months to not reached; P = .029). There was no difference in the rate of grade 3 and 4 TAEs (P = .19). In the exploratory pCSI group, 35 patients enrolled, the median CNS PFS was 5.8 months (95% CI, 4.4 to 9.1 months) and OS was 6.6 months (95% CI, 5.4 to 11 months). CONCLUSION Compared with photon IFRT, we found pCSI improved CNS PFS and OS for patients with non-small-cell lung cancer and breast cancer with LM with no increase in serious TAEs.
Collapse
Affiliation(s)
- Jonathan T. Yang
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - N. Ari Wijetunga
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Elena Pentsova
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Suzanne Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert J. Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Denise Correa
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Zhigang Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Junting Zheng
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexa Steckler
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Weronika Bucwinska
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ashley Bernstein
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Allison Betof Warner
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Melanoma and Immunotherapeutics Service, New York, NY
| | - Helena Yu
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Thoracic Oncology Service, New York, NY
| | - Mark G. Kris
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Thoracic Oncology Service, New York, NY
| | - Andrew D. Seidman
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Breast Medicine Service, New York, NY
| | - Jessica A. Wilcox
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Rachna Malani
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew Lin
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lisa M. DeAngelis
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nancy Y. Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Simon N. Powell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Adrienne Boire
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, Brain Tumor Center, New York, NY
| |
Collapse
|
24
|
Darlix A, Cayrefourcq L, Pouderoux S, Menjot de Champfleur N, Bievelez A, Jacot W, Leaha C, Thezenas S, Alix-Panabières C. Detection of Circulating Tumor Cells in Cerebrospinal Fluid of Patients with Suspected Breast Cancer Leptomeningeal Metastases: A Prospective Study. Clin Chem 2022; 68:1311-1322. [PMID: 35953885 DOI: 10.1093/clinchem/hvac127] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/30/2022] [Indexed: 11/14/2022]
Abstract
BACKGROUND The diagnosis of breast cancer (BC)-related leptomeningeal metastases (LM) relies on the detection of tumor cells in cerebrospinal fluid (CSF) using conventional cytology (gold standard). However, the sensitivity of this technique is low. Our goal was to evaluate whether circulating tumor cell (CTC) detection in CSF using the CellSearch® system could be used for LM diagnosis. METHODS This prospective, monocentric study included adult patients with suspected BC-related LM. The clinical sensitivity and specificity of CTC detection in CSF for LM diagnosis were calculated relative to conventional CSF cytology. RESULTS Forty-nine eligible patients were included and 40 were evaluable (CTC detection technical failure: n = 8, eligibility criteria failure: n = 1). Cytology was positive in 18/40 patients. CTCs were detected in these 18 patients (median: 5824 CTC, range: 93 to 45052) and in 5/22 patients with negative cytology (median: 2 CTC, range: 1 to 44). The detection of ≥1 CSF CTC was associated with a clinical sensitivity of 100% (95% CI, 82.4-100) and a specificity of 77.3% (95% CI, 64.3-90.3) for LM diagnosis. HER2+ CTCs were detected in the CSF of 40.6% of patients with HER2- BC (median: 500 CTC, range: 13 to 28 320). CONCLUSIONS The clinical sensitivity of CTC detection in CSF with the CellSearch® system for LM diagnosis is higher than that of CSF cytology. CTC detection in patients with negative cytology, however, must be further investigated. The finding of HER2+ CTCs in patients with HER2- BC suggests that the HER2 status of LM should be evaluated to increase the treatment opportunities for these patients.
Collapse
Affiliation(s)
- Amélie Darlix
- Department of Medical Oncology, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France.,Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, Montpellier, France
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, University of Montpellier, Montpellier, France.,CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
| | - Stéphane Pouderoux
- Department of Medical Oncology, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | | | - Alexis Bievelez
- Biometrics Unit, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | - William Jacot
- Department of Medical Oncology, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier IRCM, INSERM U1194, University of Montpellier; Montpellier, France
| | - Cristina Leaha
- Department of Pathology, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | - Simon Thezenas
- Biometrics Unit, Institut Régional du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, University of Montpellier, Montpellier, France.,CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
| |
Collapse
|