1
|
Bartsch R, Jerzak KJ, Larrouquere L, Müller V, Le Rhun E. Pharmacotherapy for leptomeningeal disease in breast cancer. Cancer Treat Rev 2024; 122:102653. [PMID: 38118373 DOI: 10.1016/j.ctrv.2023.102653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 12/22/2023]
Abstract
Clinical data supporting the best therapeutic approach in leptomeningeal disease (LMD; also known as leptomeningeal metastases or leptomeningeal carcinomatosis) are lacking. Despite the development of new agents and increasing incidence of central nervous system metastases, patients with LMD are often excluded from clinical trials in breast cancer, with very few conducted specifically in LMD. Consequently, current evidence may not provide an accurate reflection of real-world clinical practice. This review aims to provide further insight into the treatment strategies for patients with breast cancer and LMD. We explore differences between clinical and real-world studies, considering inclusion criteria, levels of evidence for LMD diagnosis, and time between diagnosis of LMD and LMD-specific treatment initiation. Patient prognosis is poor; median overall survival is limited to several months, with approximately 10% of patients alive at 12 months. Efficacy results have been reported for various systemic and intrathecal agents in LMD to date. Systemic therapies under investigation for LMD in breast cancer include tucatinib, trastuzumab deruxtecan, and paclitaxel trevatide; trastuzumab is the main intrathecal agent currently under investigation. Recent trials investigating systemic or intrathecal therapies are typically small, single-arm studies, and most are restricted to patients with human epidermal growth factor receptor 2-positive breast cancer. Moreover, the variability among inclusion criteria and response assessment tools makes the interpretation of results difficult. Large retrospective cohorts with various inclusion criteria and treatment regimens provide some real-world data. However, there remains an urgent need for randomised clinical trials which include patients with LMD across all breast cancer subtypes.
Collapse
Affiliation(s)
- Rupert Bartsch
- Department of Medicine I, Division of Oncology, Medical University Vienna, Vienna, Austria
| | - Katarzyna J Jerzak
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | | | - Volkmar Müller
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Emilie Le Rhun
- Departments of Neurosurgery and Neurology, University Hospital of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Bardhan M, Dey D, Suresh V, Javed B, Venur VA, Joe N, Kalidindi R, Ozair A, Khan M, Mahtani R, Lo S, Odia Y, Ahluwalia MS. An overview of the therapeutic strategies for neoplastic meningitis due to breast cancer: when and why? Expert Rev Neurother 2024; 24:77-103. [PMID: 38145503 DOI: 10.1080/14737175.2023.2293223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/06/2023] [Indexed: 12/27/2023]
Abstract
INTRODUCTION Neoplastic meningitis (NM), also known as leptomeningeal carcinomatosis, is characterized by the infiltration of tumor cells into the meninges, and poses a significant therapeutic challenge owing to its aggressive nature and limited treatment options. Breast cancer is a common cause of NM among solid tumors, further highlighting the urgent need to explore effective therapeutic strategies. This review aims to provide insights into the evolving landscape of NM therapy in breast cancer by collating existing research, evaluating current treatments, and identifying potential emerging therapeutic options. AREAS COVERED This review explores the clinical features, therapeutic strategies, recent advances, and challenges of managing NM in patients with breast cancer. Its management includes multimodal strategies, including systemic and intrathecal chemotherapy, radiation therapy, and supportive care. This review also emphasizes targeted drug options and optimal drug concentrations, and discusses emerging therapies. Additionally, it highlights the variability in treatment outcomes and the potential of combination regimens to effectively manage NM in breast cancer. EXPERT OPINION Challenges in treating NM include debates over clinical trial end points and the management of adverse effects. Drug resistance and low response rates are significant hurdles, particularly inHER2-negative breast cancer. The development of more precise and cost-effective medications with improved selectivity is crucial. Additionally, global efforts are needed for infrastructure development and cancer control considering the diverse nature of the disease.
Collapse
Affiliation(s)
- Mainak Bardhan
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | | | - Vinay Suresh
- King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Binish Javed
- Atal Bihari Vajpayee Institute of Medical Sciences and Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Vyshak Alva Venur
- Seattle Cancer Care Alliance, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Neha Joe
- St John's Medical College Hospital, Bengaluru, India
| | | | - Ahmad Ozair
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Reshma Mahtani
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Simon Lo
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, USA
| | - Yazmin Odia
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Manmeet S Ahluwalia
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
3
|
Ghanem P, Fatteh M, Kamson DO, Balan A, Chang M, Tao J, Blakeley J, Canzoniero J, Grossman SA, Marrone K, Schreck KC, Anagnostou V. Druggable genomic landscapes of high-grade gliomas. Front Med (Lausanne) 2023; 10:1254955. [PMID: 38143440 PMCID: PMC10749203 DOI: 10.3389/fmed.2023.1254955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/06/2023] [Indexed: 12/26/2023] Open
Abstract
Background Despite the putatively targetable genomic landscape of high-grade gliomas, the long-term survival benefit of genomically-tailored targeted therapies remains discouraging. Methods Using glioblastoma (GBM) as a representative example of high-grade gliomas, we evaluated the clonal architecture and distribution of hotspot mutations in 388 GBMs from the Cancer Genome Atlas (TCGA). Mutations were matched with 54 targeted therapies, followed by a comprehensive evaluation of drug biochemical properties in reference to the drug's clinical efficacy in high-grade gliomas. We then assessed clinical outcomes of a cohort of patients with high-grade gliomas with targetable mutations reviewed at the Johns Hopkins Molecular Tumor Board (JH MTB; n = 50). Results Among 1,156 sequence alterations evaluated, 28.6% represented hotspots. While the frequency of hotspot mutations in GBM was comparable to cancer types with actionable hotspot alterations, GBMs harbored a higher fraction of subclonal mutations that affected hotspots (7.0%), compared to breast cancer (4.9%), lung cancer (4.4%), and melanoma (1.4%). In investigating the biochemical features of targeted therapies paired with recurring alterations, we identified a trend toward higher lipid solubility and lower IC50 in GBM cell lines among drugs with clinical efficacy. The drugs' half-life, molecular weight, surface area and binding to efflux transporters were not associated with clinical efficacy. Among the JH MTB cohort of patients with IDH1 wild-type high-grade gliomas who received targeted therapies, trametinib monotherapy or in combination with dabrafenib conferred radiographic partial response in 75% of patients harboring BRAF or NF1 actionable mutations. Cabozantinib conferred radiographic partial response in two patients harboring a MET and a PDGFRA/KDR amplification. Patients with IDH1 wild-type gliomas that harbored actionable alterations who received genotype-matched targeted therapy had longer progression-free (PFS) and overall survival (OS; 7.37 and 14.72 respectively) than patients whose actionable alterations were not targeted (2.83 and 4.2 months respectively). Conclusion While multiple host, tumor and drug-related features may limit the delivery and efficacy of targeted therapies for patients with high-grade gliomas, genotype-matched targeted therapies confer favorable clinical outcomes. Further studies are needed to generate more data on the impact of biochemical features of targeted therapies on their clinical efficacy for high-grade gliomas.
Collapse
Affiliation(s)
- Paola Ghanem
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Johns Hopkins Molecular Tumor Board, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Maria Fatteh
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Johns Hopkins Molecular Tumor Board, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David Olayinka Kamson
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Archana Balan
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Johns Hopkins Molecular Tumor Board, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael Chang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jessica Tao
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Johns Hopkins Molecular Tumor Board, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jaishri Blakeley
- The Johns Hopkins Molecular Tumor Board, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jenna Canzoniero
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Johns Hopkins Molecular Tumor Board, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Stuart A. Grossman
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Johns Hopkins Molecular Tumor Board, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kristen Marrone
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Karisa C. Schreck
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Johns Hopkins Molecular Tumor Board, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Valsamo Anagnostou
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Johns Hopkins Molecular Tumor Board, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
Puszkiel A, Bousquet G, Stanke-Labesque F, Stocco J, Decq P, Chevillard L, Goutagny S, Declèves X. A Minimal PBPK Model for Plasma and Cerebrospinal Fluid Pharmacokinetics of Trastuzumab after Intracerebroventricular Administration in Patients with HER2-Positive Brain Metastatic Localizations. Pharm Res 2023; 40:2687-2697. [PMID: 37821769 DOI: 10.1007/s11095-023-03614-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Dosing regimens of trastuzumab administered by intracerebroventricular (icv) route to patients with HER2-positive brain localizations remain empirical. The objectives of this study were to describe pharmacokinetics (PK) of trastuzumab in human plasma and cerebrospinal fluid (CSF) after simultaneous icv and intravenous (iv) administration using a minimal physiologically-based pharmacokinetic model (mPBPK) and to perform simulations of alternative dosing regimens to achieve therapeutic concentrations in CSF. METHODS Plasma and CSF PK data were collected in two patients with HER2-positive brain localizations. A mPBPK model for mAbs consisting of four compartments (tight and leaky tissues, plasma and lymph) was enriched by an additional compartment for ventricular CSF. The comparison between observed and model-predicted concentrations was evaluated using prediction error (PE). RESULTS The developed mPBPK model described plasma and CSF trastuzumab concentrations reasonably well with mean PE for plasma and CSF data of 41.8% [interquartile range, IQR = -9.48; 40.6] and 18.3% [-36.7; 60.6], respectively, for patient 1 and 11.4% [-10.8; 28.7] and 22.5% [-27.7; 77.9], respectively, for patient 2. Trastuzumab showed fast clearance from CSF to plasma with Cmin,ss of 0.56 and 0.85 mg/L for 100 and 150 mg q1wk, respectively. Repeated dosing of 100 and 150 mg q3day resulted in Cmin,ss of 10.3 and 15.4 mg/L, respectively. Trastuzumab CSF target concentrations are achieved rapidly and maintained above 60 mg/L from 7 days after a continuous perfusion at 1.0 mg/h. CONCLUSION Continuous icv infusion of trastuzumab at 1.0 mg/h could be an alternative dosing regimen to rapidly achieve intraventricular CSF therapeutic concentrations.
Collapse
Affiliation(s)
- Alicja Puszkiel
- Université Paris Cité, Inserm UMRS1144, Paris, France
- Laboratory of Pharmacology and Toxicology, Cochin University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Guilhem Bousquet
- Oncology Department, Avicenne Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
- Université Paris Cité, Inserm UMR_S942 MASCOT, Paris, France
- Université Sorbonne Paris Nord, Villetaneuse, France
| | - Françoise Stanke-Labesque
- Laboratory of Pharmacology, Toxicology and Pharmacogenetics, Grenoble-Alpes University Hospital, 38043, Grenoble, France
- Université Grenoble Alpes, HP2 INSERM U1300, Grenoble, France
| | - Jeanick Stocco
- Department of Pharmacy, Beaujon Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Philippe Decq
- Department of Neurosurgery, Beaujon University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | - Stéphane Goutagny
- Université Paris Cité, Inserm UMRS1144, Paris, France
- Department of Neurosurgery, Beaujon University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Xavier Declèves
- Université Paris Cité, Inserm UMRS1144, Paris, France.
- Laboratory of Pharmacology and Toxicology, Cochin University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France.
| |
Collapse
|
5
|
Lazaratos AM, Maritan SM, Quaiattini A, Darlix A, Ratosa I, Ferraro E, Griguolo G, Guarneri V, Pellerino A, Hofer S, Jacot W, Stemmler HJ, van den Broek MPH, Dobnikar N, Panet F, Lahijanian Z, Morikawa A, Seidman AD, Soffietti R, Panasci L, Petrecca K, Rose AAN, Bouganim N, Dankner M. Intrathecal trastuzumab versus alternate routes of delivery for HER2-targeted therapies in patients with HER2+ breast cancer leptomeningeal metastases. Breast 2023; 69:451-468. [PMID: 37156650 PMCID: PMC10300571 DOI: 10.1016/j.breast.2023.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/07/2023] [Accepted: 04/30/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Patients with HER2+ breast cancer (BC) frequently develop leptomeningeal metastases (LM). While HER2-targeted therapies have demonstrated efficacy in the neoadjuvant, adjuvant, and metastatic settings, including for parenchymal brain metastases, their efficacy for patients with LM has not been studied in a randomized controlled trial. However, several single-armed prospective studies, case series and case reports have studied oral, intravenous, or intrathecally administered HER2-targeted therapy regimens for patients with HER2+ BC LM. METHODS We conducted a systematic review and meta-analysis of individual patient data to evaluate the efficacy of HER2-targeted therapies in HER2+ BC LM in accordance with PRISMA guidelines. Targeted therapies evaluated were trastuzumab (intrathecal or intravenous), pertuzumab, lapatinib, neratinib, tucatinib, trastuzumab-emtansine and trastuzumab-deruxtecan. The primary endpoint was overall survival (OS), with CNS-specific progression-free survival (PFS) as a secondary endpoint. RESULTS 7780 abstracts were screened, identifying 45 publications with 208 patients, corresponding to 275 lines of HER2-targeted therapy for BC LM which met inclusion criteria. In univariable and multivariable analyses, we observed no significant difference in OS and CNS-specific PFS between intrathecal trastuzumab compared to oral or intravenous administration of HER2-targeted therapy. Anti-HER2 monoclonal antibody-based regimens did not demonstrate superiority over HER2 tyrosine kinase inhibitors. In a cohort of 15 patients, treatment with trastuzumab-deruxtecan was associated with prolonged OS compared to other HER2-targeted therapies and compared to trastuzumab-emtansine. CONCLUSIONS The results of this meta-analysis, comprising the limited data available, suggest that intrathecal administration of HER2-targeted therapy for patients with HER2+ BC LM confers no additional benefit over oral and/or IV treatment regimens. Although the number of patients receiving trastuzumab deruxtecan in this cohort is small, this novel agent offers promise for this patient population and requires further investigation in prospective studies.
Collapse
Affiliation(s)
- Anna-Maria Lazaratos
- Rosalind and Morris Goodman Cancer Institute, Montreal, Quebec, Canada; Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Sarah M Maritan
- Rosalind and Morris Goodman Cancer Institute, Montreal, Quebec, Canada; Faculty of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Andrea Quaiattini
- Schulich Library of Physical Sciences, Life Sciences, and Engineering, McGill University, Montreal, Quebec, Canada
| | - Amelie 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
| | - Ivica Ratosa
- Division of Radiotherapy, Institute of Oncology Ljubljana, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Emanuela Ferraro
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, NewYork, USA
| | - Gaia Griguolo
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Valentina Guarneri
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Alessia Pellerino
- Division of Neuro-Oncology, Department of Neuroscience, University and City of Health and Science Hospital, Turin, Italy
| | - Silvia Hofer
- Department of Neurology, University Hospital Zurich, Switzerland
| | - William Jacot
- Department of Medical Oncology, Institut régional du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | | | | | - Nika Dobnikar
- Division of Radiotherapy, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Francois Panet
- Gerald Bronfman Department of Oncology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Zubin Lahijanian
- Department of Diagnostic Radiology, McGill University, Montreal, Quebec, Canada
| | - Aki Morikawa
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, NewYork, USA
| | - Andrew D Seidman
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, NewYork, USA
| | - Riccardo Soffietti
- Division of Neuro-Oncology, Department of Neuroscience, University and City of Health and Science Hospital, Turin, Italy
| | - Lawrence Panasci
- Gerald Bronfman Department of Oncology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Kevin Petrecca
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - April A N Rose
- Gerald Bronfman Department of Oncology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada; Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Nathaniel Bouganim
- Gerald Bronfman Department of Oncology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada; McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Matthew Dankner
- Rosalind and Morris Goodman Cancer Institute, Montreal, Quebec, Canada; Faculty of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
6
|
Oberkampf F, Gutierrez M, Trabelsi Grati O, Rhun ÉL, Trédan O, Turbiez I, Kadi A, Dubot C, Taillibert S, Vacher S, Bonneau C. Phase II study of intrathecal administration of trastuzumab in patients with HER2-positive breast cancer with leptomeningeal metastasis. Neuro Oncol 2022; 25:365-374. [PMID: 35868630 PMCID: PMC9925703 DOI: 10.1093/neuonc/noac180] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Patients with HER2-positive breast cancer (HER2 + BC) develop central nervous system metastases twice as often as patients with luminal HER2-negative breast cancer. Leptomeningeal progression results in a drastically altered prognosis with limited therapeutic options. This phase II study was conducted to assess the efficacy of intrathecal (IT) trastuzumab in HER2 + BC patients with leptomeningeal metastasis (LM), based on a phase I dose-escalation study that had determined the recommended weekly dose of 150 mg for phase II. METHODS Eligible patients received weekly administrations of 150 mg of IT trastuzumab. The primary endpoint was clinical neurologic progression-free survival (LM-related-PFS) after 8 weeks. Overall survival (OS), toxicities, and quality of life (QoL) were secondary endpoints. RESULTS Among the 19 enrolled patients, 16 (84%) had concomitant brain metastases, 15 of them had received prior radiotherapy to the brain. All patients had received at least one line of systemic anti-HER2 therapy. After 8 weeks, 14 patients (74%) were free of neurological progression. The median LM-related-PFS was 5.9 months and the median OS was 7.9 months. According to the QLQ-C30 and BN20 scales, the global health-related QoL status seemed preserved and no toxicity above grade 3 was observed. CONCLUSIONS Conducting studies on patients with LM poses significant challenges and ethical dilemmas inherent to this population. Despite some limits, this phase II study's findings in terms of clinical neurologic response and QoL's control confirms weekly administration of 150 mg of IT trastuzumab as a valuable option for HER + BC patients with LM and support the interest for further investigations.
Collapse
Affiliation(s)
- Florence Oberkampf
- Department of Oncology, Institut Curie-St Cloud, 92210, Saint Cloud, France
| | - Maya Gutierrez
- Department of Oncology, Institut Curie-St Cloud, 92210, Saint Cloud, France
| | | | - Émilie Le Rhun
- Neuro-Oncology Neurosurgery Department, University of LilleFrance, CHU Lille, France,Neurology, Department of Medical Oncology, Centre Oscar Lambret, Lille, France
| | - Olivier Trédan
- Department of Oncology, Centre Leon Berard, 69008, Lyon, France
| | - Isabelle Turbiez
- Department of Clinical Research, Institut Curie-St Cloud, 92210, Saint Cloud, France
| | - Amir Kadi
- Department of Biostatistics, Institut Curie-St Cloud, 92210, Saint Cloud, France
| | - Coraline Dubot
- Department of Oncology, Institut Curie-St Cloud, 92210, Saint Cloud, France
| | - Sophie Taillibert
- Department of Neurology Mazarin, Groupe Hospitalier Pitié-Salpêtrière, 75013, Paris, France
| | - Sophie Vacher
- Department of Genetics, Institut Curie-Paris, 75005, Paris, France
| | - Claire Bonneau
- Corresponding Author: Claire Bonneau, Department of surgery, Institut Curie-Saint-Cloud, 35 Rue Dailly, 92210 Saint Cloud, France ()
| |
Collapse
|