1
|
Li Y, Liu F, Cai Q, Deng L, Ouyang Q, Zhang XHF, Zheng J. Invasion and metastasis in cancer: molecular insights and therapeutic targets. Signal Transduct Target Ther 2025; 10:57. [PMID: 39979279 PMCID: PMC11842613 DOI: 10.1038/s41392-025-02148-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
The progression of malignant tumors leads to the development of secondary tumors in various organs, including bones, the brain, liver, and lungs. This metastatic process severely impacts the prognosis of patients, significantly affecting their quality of life and survival rates. Research efforts have consistently focused on the intricate mechanisms underlying this process and the corresponding clinical management strategies. Consequently, a comprehensive understanding of the biological foundations of tumor metastasis, identification of pivotal signaling pathways, and systematic evaluation of existing and emerging therapeutic strategies are paramount to enhancing the overall diagnostic and treatment capabilities for metastatic tumors. However, current research is primarily focused on metastasis within specific cancer types, leaving significant gaps in our understanding of the complex metastatic cascade, organ-specific tropism mechanisms, and the development of targeted treatments. In this study, we examine the sequential processes of tumor metastasis, elucidate the underlying mechanisms driving organ-tropic metastasis, and systematically analyze therapeutic strategies for metastatic tumors, including those tailored to specific organ involvement. Subsequently, we synthesize the most recent advances in emerging therapeutic technologies for tumor metastasis and analyze the challenges and opportunities encountered in clinical research pertaining to bone metastasis. Our objective is to offer insights that can inform future research and clinical practice in this crucial field.
Collapse
Affiliation(s)
- Yongxing Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Graduate School of Biomedical Science, Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lijun Deng
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
2
|
Grinda T, Aizer AA, Lin NU, Sammons SL. Central Nervous System Metastases in Breast Cancer. Curr Treat Options Oncol 2025; 26:14-35. [PMID: 39786689 DOI: 10.1007/s11864-024-01286-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2024] [Indexed: 01/12/2025]
Abstract
OPINION STATEMENT Breast cancer metastasizing to the central nervous system (CNS) encompasses two distinct entities: brain metastases involving the cerebral parenchyma and infiltration of the leptomeningeal space, i.e., leptomeningeal disease. CNS metastases affect 10-15% of patients with hormone receptor-positive-status and nearly one-half of those with HER2-positive and triple-negative breast cancer with distant metastatic disease. Significant clinical morbidity and heterogeneous penetration of the blood-brain barrier by systemic therapies contribute to the poor prognosis associated with brain metastases. Recent advances in radiotherapy, including stereotactic approaches and morbidity-reducing strategies such as the use of memantine and hippocampal avoidance in whole brain radiation, coupled with the development of more effective CNS-penetrant systemic therapies, including small molecules and antibody-drug conjugates, have significantly improved patient outcomes. Consequently, patients with breast cancer CNS metastases have improved survival compared to prior decades, and longitudinal care has become increasingly complex, necessitating a multidisciplinary approach to achieve optimal outcomes for patients.
Collapse
Affiliation(s)
- Thomas Grinda
- Breast Oncology Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Harvard Medical School, Boston, MA, USA
| | - Ayal A Aizer
- Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nancy U Lin
- Breast Oncology Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sarah L Sammons
- Breast Oncology Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Podder V, Bellur S, Margolin K, Advani P, Mahtani RL, Subbiah V, Novo GB, Ranjan T, Ahluwalia MS. Immunotherapeutic and Targeted Strategies for Managing Brain Metastases from Common Cancer Origins: A State-of-the-Art Review. Curr Oncol Rep 2024; 26:1612-1638. [PMID: 39514054 DOI: 10.1007/s11912-024-01593-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW This review examines contemporary strategies for managing brain metastases (BM) from common cancers such as lung, breast, and melanoma. We evaluate the efficacy and applicability of targeted therapies and immunotherapies, exploring their potential to cross the blood-brain barrier and improve patient outcomes. RECENT FINDINGS Recent studies have shown that tyrosine kinase inhibitors, immune checkpoint inhibitors, and ADCs effectively treat BM. These treatments can overcome the challenges posed by the blood-brain barrier and improve therapeutic outcomes. ADCs are promising because they can deliver cytotoxic agents directly to tumor cells, which reduces systemic toxicity and increases drug delivery efficiency to the brain. Personalized medicine is becoming increasingly significant in treatment decisions, with biomarkers playing an essential role. Advances in molecular genetics and drug development have led to more refined treatments, emphasizing the precision medicine framework. The management of BM is evolving, driven by drug efficacy, resistance mechanisms, and the need for personalized medicine. Integrating ADCs into treatment regimens represents a significant advancement in targeting metastatic brain tumors. Despite these advances, BM management still presents considerable challenges, requiring ongoing research and multi-institutional trials to optimize therapeutic strategies. This review outlines the current state and future directions in treating BM, highlighting the critical need for continued innovation and comprehensive clinical evaluations to improve survival rates and quality of life for affected patients.
Collapse
Affiliation(s)
- Vivek Podder
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Shreyas Bellur
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Kim Margolin
- Saint John's Cancer Institute, Santa Monica, CA, USA
| | | | - Reshma L Mahtani
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Vivek Subbiah
- Sarah Cannon Research Institute (SCRI), Nashville, TN, USA
| | - Gabriella B Novo
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Tulika Ranjan
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | | |
Collapse
|
4
|
Silvestri VL, Tran AD, Chung M, Chung N, Gril B, Robinson C, Difilippantonio S, Wei D, Kruhlak MJ, Peer CJ, Figg WD, Khan I, Steeg PS. Distinct uptake and elimination profiles for trastuzumab, human IgG, and biocytin-TMR in experimental HER2+ brain metastases of breast cancer. Neuro Oncol 2024; 26:1067-1082. [PMID: 38363979 PMCID: PMC11145443 DOI: 10.1093/neuonc/noae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND The aim of this study is an improved understanding of drug distribution in brain metastases. Rather than single point snapshots, we analyzed the time course and route of drug/probe elimination (clearance), focusing on the intramural periarterial drainage (IPAD) pathway. METHODS Mice with JIMT1-BR HER2+ experimental brain metastases were injected with biocytin-TMR and either trastuzumab or human IgG. Drugs/probes circulated for 5 min to 48 h, followed by perfusion. Brain sections were stained for human IgG, vascular basement membrane proteins laminin or collagen IV, and periarterial α-SMA. A machine learning algorithm was developed to identify metastases, metastatic microenvironment, and uninvolved brain in confocally scanned brain sections. Drug/probe intensity over time and total imaged drug exposure (iAUC) were calculated for 27,249 lesions and co-immunofluorescence with IPAD-vascular matrix analyzed in 11,668 metastases. RESULTS In metastases, peak trastuzumab levels were 5-fold higher than human IgG but 4-fold less than biocytin-TMR. The elimination phase constituted 85-93% of total iAUC for all drugs/probes tested. For trastuzumab, total iAUC during uptake was similar to the small molecule drug probe biocytin-TMR, but slower trastuzumab elimination resulted in a 1.7-fold higher total iAUC. During elimination trastuzumab and IgG were preferentially enriched in the α-SMA+ periarterial vascular matrix, consistent with the IPAD clearance route; biocytin-TMR showed heterogeneous elimination pathways. CONCLUSIONS Drug/probe elimination is an important component of drug development for brain metastases. We identified a prolonged elimination pathway for systemically administered antibodies through the periarterial vascular matrix that may contribute to the sustained presence and efficacy of large antibody therapeutics.
Collapse
Affiliation(s)
- Vanesa L Silvestri
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Andy D Tran
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
- CCR Microscopy Core, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Monika Chung
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Natalie Chung
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Brunilde Gril
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Christina Robinson
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Debbie Wei
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Michael J Kruhlak
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
- CCR Microscopy Core, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Cody J Peer
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - W Douglas Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Imran Khan
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Patricia S Steeg
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Xu D, Hu Z, Wang K, Hu S, Zhou Y, Zhang S, Chen Y, Pan T. Why does HER2-positive breast cancer metastasize to the brain and what can we do about it? Crit Rev Oncol Hematol 2024; 195:104269. [PMID: 38272149 DOI: 10.1016/j.critrevonc.2024.104269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 12/18/2023] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Breast cancer is the most frequent malignancy in women. However, in the middle and late stages, some people develop distant metastases, which considerably lower the quality of life and life expectancy. The brain is one of the sites where metastasis frequently happens. According to epidemiological research, brain metastases occur at a late stage in 30-50% of patients with HER2-positive breast cancer, resulting in a poor prognosis. Additionally, few treatments are available for HER2-positive brain metastatic breast cancer, and the mortality rate is remarkable owing to the complexity of the brain's anatomical structure and physiological function. In this review, we described the stages of the brain metastasis of breast cancer, the relationship between the microenvironment and metastatic cancer cells, and the unique molecular and cellular mechanisms. It involves cancer cells migrating, invading, and adhering to the brain; penetrating the blood-brain barrier; interacting with brain cells; and activating signal pathways once inside the brain. Finally, we reviewed current clinically used treatment approaches for brain metastasis in HER2-positive breast cancer; summarized the traditional treatment, targeted treatment, immunotherapy, and other treatment modalities; compared the benefits and drawbacks of each approach; discussed treatment challenges; and emphasized the importance of identifying potential targets to improve patient survival rates and comprehend brain metastasis in breast cancer.
Collapse
Affiliation(s)
- Dongyan Xu
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Zhengfang Hu
- Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China
| | - Kaiyue Wang
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shiyao Hu
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yunxiang Zhou
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shizhen Zhang
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yiding Chen
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Tao Pan
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
6
|
Liang X, Gui X, Yan Y, Di L, Liu X, Li H, Song G. Long-term Outcome Analysis of Pyrotinib in Patients With HER2-Positive Metastatic Breast Cancer and Brain Metastasis: A Real-World Study. Oncologist 2024; 29:e198-e205. [PMID: 37589217 PMCID: PMC10836298 DOI: 10.1093/oncolo/oyad228] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/19/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Pyrotinib is currently approved for the treatment of HER2-positive advanced breast cancer in China. Data on the overall survival (OS) and efficacy in patients with brain metastasis (BM) remain scarce. This study evaluated the effectiveness of pyrotinib in a real-world setting, especially in patients with BM. METHODS We reviewed patients with metastatic breast cancer treated with pyrotinib-based therapy between June 2018 and June 2022. Progression-free survival (PFS), OS, objective response rate, and safety were analyzed following the administration of pyrotinib. RESULTS A total of 239 patients were included. The median PFS in patients who received pyrotinib-based therapy as first-line (15/239), second-line (115/239), or third-or-higher-line (109/239) treatment was 14.00, 9.33, and 8.20 months, respectively, and the median OS was not reached, 29.07 and 22.23 months, respectively. The median PFS in patients who pretreated with trastuzumab (214/239), trastuzumab plus pertuzumab (22/239), lapatinib (68/239), or trastuzumab emtansine (14/239) was 9.33, 6.87, 7.20, and 7.20 months, respectively. In 61 patients with BM, the median PFS was 7.50 months, the median central nervous system (CNS)-PFS was 11.17 months, and the median OS was 21.27 months. Furthermore, 19 patients with concomitant brain radiotherapy tended to achieve a longer OS than 42 patients without radiation (34.17 vs. 20.70 months, P = .112). CONCLUSIONS Long-term outcomes of pyrotinib-based therapy are promising for patients with HER2-positive metastatic breast cancer in real world and in patients with BM, regardless of the treatment lines and prior anti-HER2 therapies.
Collapse
Affiliation(s)
- Xu Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| | - Xinyu Gui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| | - Ying Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| | - Lijun Di
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| | - Xiaoran Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| | - Huiping Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| | - Guohong Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| |
Collapse
|
7
|
Buczek D, Zaucha R, Jassem J. Neurotoxicity-sparing radiotherapy for brain metastases in breast cancer: a narrative review. Front Oncol 2024; 13:1215426. [PMID: 38370347 PMCID: PMC10869626 DOI: 10.3389/fonc.2023.1215426] [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: 05/01/2023] [Accepted: 12/19/2023] [Indexed: 02/20/2024] Open
Abstract
Breast cancer brain metastasis (BCBM) has a devastating impact on patient survival, cognitive function and quality of life. Radiotherapy remains the standard management of BM but may result in considerable neurotoxicity. Herein, we describe the current knowledge on methods for reducing radiation-induced cognitive dysfunction in patients with BCBM. A better understanding of the biology and molecular underpinnings of BCBM, as well as more sophisticated prognostic models and individualized treatment approaches, have appeared to enable more effective neuroprotection. The therapeutic armamentarium has expanded from surgery and whole-brain radiotherapy to stereotactic radiosurgery, targeted therapies and immunotherapies, used sequentially or in combination. Advances in neuroimaging have allowed more accurate screening for intracranial metastases, precise targeting of intracranial lesions and the differentiation of the effects of treatment from disease progression. The availability of numerous treatment options for patients with BCBM and multidisciplinary approaches have led to personalized treatment and improved therapeutic outcomes. Ongoing studies may define the optimal sequencing of available and emerging treatment options for patients with BCBM.
Collapse
|
8
|
Caputo R, Buono G, Di Lauro V, Cianniello D, Von Arx C, Pensabene M, Pagliuca M, Pacilio C, Di Rella F, Verrazzo A, Martinelli C, Nuzzo F, De Laurentiis M. Neratinib as adjuvant therapy in patients with HER2 positive breast cancer: expert opinion. Future Oncol 2023; 19:1695-1708. [PMID: 37605877 DOI: 10.2217/fon-2023-0361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/19/2023] [Indexed: 08/23/2023] Open
Abstract
Neratinib is a tyrosine kinase receptor inhibitor used in the extended adjuvant therapy of early-stage breast cancer. After adjuvant trastuzumab therapy, neratinib reduces the risk of recurrence and, if taken within 1 year from trastuzumab, significantly improves the invasive disease-free survival of patients with early-stage human epidermal growth factor receptor-2 positive (HER2+) breast cancer with no increased risk of long-term toxicity. Diarrhea, the most common adverse event associated with neratinib use, deters some clinicians from prescribing this drug. However, neratinib-related toxicity is predictable, short-lived, mostly limited to the first month of treatment and can be managed with dose-escalation and prophylactic strategies. Thus, close surveillance and prompt management, relying on supportive care and administration schedule modification, allows discontinuation of treatment to be avoided.
Collapse
Affiliation(s)
- Roberta Caputo
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| | - Giuseppe Buono
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| | - Vincenzo Di Lauro
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| | - Daniela Cianniello
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| | - Claudia Von Arx
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| | - Matilde Pensabene
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| | - Martina Pagliuca
- Clinical & Translational Oncology, Scuola Superiore Meridionale, Naples, Italy
- U981 Molecular Predictors & New Targets in Oncology, Gustave Roussy, Villejuif, France
| | - Carmen Pacilio
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| | - Francesca Di Rella
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| | - Annarita Verrazzo
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
- Clinical & Translational Oncology, Scuola Superiore Meridionale, Naples, Italy
- Department of Clinical Medicine & Surgery, University of Naples Federico II, Naples, Italy
| | - Claudia Martinelli
- Department of Clinical Medicine & Surgery, University of Naples Federico II, Naples, Italy
| | - Francesco Nuzzo
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| | - Michelino De Laurentiis
- Department of Breast & Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale,' Naples, Italy
| |
Collapse
|
9
|
Roy-O'Reilly MA, Lanman T, Ruiz A, Rogawski D, Stocksdale B, Nagpal S. Diagnostic and Therapeutic Updates in Leptomeningeal Disease. Curr Oncol Rep 2023; 25:937-950. [PMID: 37256537 PMCID: PMC10326117 DOI: 10.1007/s11912-023-01432-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/01/2023]
Abstract
PURPOSE OF REVIEW Leptomeningeal disease (LMD) is a devastating complication of advanced metastatic cancer associated with a poor prognosis and limited treatment options. This study reviews the current understanding of the clinical presentation, pathogenesis, diagnosis, and treatment of LMD. We highlight opportunities for advances in this disease. RECENT FINDINGS In recent years, the use of soluble CSF biomarkers has expanded, suggesting improved sensitivity over traditional cytology, identification of targetable mutations, and potential utility for monitoring disease burden. Recent studies of targeted small molecules and intrathecal based therapies have demonstrated an increase in overall and progression-free survival. In addition, there are several ongoing trials evaluating immunotherapy in LMD. Though overall prognosis of LMD remains poor, studies suggest a potential role for soluble CSF biomarkers in diagnosis and management and demonstrate promising findings in patient outcomes with targeted therapies for specific solid tumors. Despite these advances, there continues to be a gap of knowledge in this disease, emphasizing the importance of inclusion of LMD patients in clinical trials.
Collapse
Affiliation(s)
| | - Tyler Lanman
- Department of Neurology, Stanford Medicine, Palo Alto, CA, 94305, USA
| | - Amber Ruiz
- Department of Neurology, Stanford Medicine, Palo Alto, CA, 94305, USA
| | - David Rogawski
- Department of Neurology, Stanford Medicine, Palo Alto, CA, 94305, USA
| | - Brian Stocksdale
- Department of Neurology, Stanford Medicine, Palo Alto, CA, 94305, USA
| | - Seema Nagpal
- Department of Neurology, Stanford Medicine, Palo Alto, CA, 94305, USA.
| |
Collapse
|
10
|
Kioutchoukova I, Lucke-Wold BP. Pyrotinib as a therapeutic for HER2-positive breast cancer. Transl Cancer Res 2023; 12:1376-1379. [PMID: 37434682 PMCID: PMC10331451 DOI: 10.21037/tcr-23-333] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/19/2023] [Indexed: 07/13/2023]
|
11
|
Jenkins S, Zhang W, Steinberg SM, Nousome D, Houston N, Wu X, Armstrong TS, Burton E, Smart DD, Shah R, Peer CJ, Mozarsky B, Arisa O, Figg WD, Mendoza TR, Vera E, Brastianos P, Carter S, Gilbert MR, Anders CK, Connolly RM, Tweed C, Smith KL, Khan I, Lipkowitz S, Steeg PS, Zimmer AS. Phase I Study and Cell-Free DNA Analysis of T-DM1 and Metronomic Temozolomide for Secondary Prevention of HER2-Positive Breast Cancer Brain Metastases. Clin Cancer Res 2023; 29:1450-1459. [PMID: 36705597 PMCID: PMC10153633 DOI: 10.1158/1078-0432.ccr-22-0855] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 11/22/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023]
Abstract
PURPOSE Preclinical data showed that prophylactic, low-dose temozolomide (TMZ) significantly prevented breast cancer brain metastasis. We present results of a phase I trial combining T-DM1 with TMZ for the prevention of additional brain metastases after previous occurrence and local treatment in patients with HER2+ breast cancer. PATIENTS AND METHODS Eligible patients had HER2+ breast cancer with brain metastases and were within 12 weeks of whole brain radiation therapy (WBRT), stereotactic radiosurgery, and/or surgery. Standard doses of T-DM1 were administered intravenously every 21 days (3.6 mg/kg) and TMZ was given orally daily in a 3+3 phase I dose escalation design at 30, 40, or 50 mg/m2, continuously. DLT period was one 21-day cycle. Primary endpoint was safety and recommended phase II dose. Symptom questionnaires, brain MRI, and systemic CT scans were performed every 6 weeks. Cell-free DNA sequencing was performed on patients' plasma and CSF. RESULTS Twelve women enrolled, nine (75%) with prior SRS therapy and three (25%) with prior WBRT. Grade 3 or 4 AEs included thrombocytopenia (1/12), neutropenia (1/12), lymphopenia (6/12), and decreased CD4 (6/12), requiring pentamidine for Pneumocystis jirovecii pneumonia prophylaxis. No DLT was observed. Four patients on the highest TMZ dose underwent dose reductions. At trial entry, 6 of 12 patients had tumor mutations in CSF, indicating ongoing metastatic colonization despite a clear MRI. Median follow-up on study was 9.6 m (2.8-33.9); only 2 patients developed new parenchymal brain metastases. Tumor mutations varied with patient outcome. CONCLUSIONS Metronomic TMZ in combination with standard dose T-DM1 shows low-grade toxicity and potential activity in secondary prevention of HER2+ brain metastases.
Collapse
Affiliation(s)
- Sarah Jenkins
- Women’s Malignancies Branch; Center for Cancer Research, NCI, NIH
| | - Wei Zhang
- Women’s Malignancies Branch; Center for Cancer Research, NCI, NIH
| | - Seth M. Steinberg
- Biostatistics and Data Management Section; Center for Cancer Research, NCI, NIH
| | - Darryl Nousome
- Center for Cancer Research Collaborative Bioinformatics Resource, NCI, NIH
| | - Nicole Houston
- Women’s Malignancies Branch; Center for Cancer Research, NCI, NIH
| | - Xiaolin Wu
- Cancer Research Technology Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | | | - Dee Dee Smart
- Radiation Oncology Branch, Center for Cancer Research, NCI NIH
| | - Ritu Shah
- Neuro-Radiology, Clinical Center Cancer Research, NIH
| | - Cody J. Peer
- Clinical Pharmacology Program, Center for Cancer Research, NCI NIH
| | - Brett Mozarsky
- Clinical Pharmacology Program, Center for Cancer Research, NCI NIH
| | - Oluwatobi Arisa
- Clinical Pharmacology Program, Center for Cancer Research, NCI NIH
| | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, NCI NIH
| | | | | | - Priscilla Brastianos
- Massachusetts General Hospital, Harvard Cancer Center, Harvard University, Boston, MA
| | - Scott Carter
- Division of Medical Sciences, Harvard University, Boston, MA
| | | | | | | | - Carol Tweed
- University of Maryland Oncology, Baltimore MD
| | - Karen L. Smith
- Johns Hopkins University School of Medicine, Baltimore, MD
| | - Imran Khan
- Women’s Malignancies Branch; Center for Cancer Research, NCI, NIH
| | | | | | | |
Collapse
|
12
|
Mandó P, Waisberg F, Pasquinelli R, Rivero S, Ostinelli A, Perazzo F. HER2-Directed Therapy in Advanced Breast Cancer: Benefits and Risks. Onco Targets Ther 2023; 16:115-132. [PMID: 36844609 PMCID: PMC9948634 DOI: 10.2147/ott.s335934] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/20/2023] [Indexed: 02/20/2023] Open
Abstract
Around 20% of breast cancers are associated with amplification or overexpression of human epidermal growth factor receptor 2 (HER2). In this setting, anti-HER2-targeted agents are the cornerstone of cancer therapeutic strategies. This includes monoclonal antibodies, tyrosine kinase inhibitors (TKIs) and, recently, antibody-drug conjugates (ADCs). With the advent of these new alternatives, the decision-making process has become more complex, especially with regard to the treatment sequence possibilities. In spite of the fact that overall survival has significantly improved accordingly, resistance to treatment remains a challenge in HER2-positive breast cancer. The introduction of new agents has created awareness regarding new potential specific adverse events, and consequently, their increasing application pose major challenges in daily patient care. This review describes the therapeutic landscape for HER2-positive advanced breast cancer (ABC) and evaluates its benefits and risks in the clinical setting.
Collapse
Affiliation(s)
- Pablo Mandó
- Clinical Oncology Department, Centro de Educación Médica e Investigaciones Clínicas “Norberto Quirno” (CEMIC), Ciudad Autónoma de Buenos Aires, Argentina
| | - Federico Waisberg
- Clinical Oncology Department, Instituto Alexander Fleming, Ciudad Autónoma de Buenos Aires, Argentina
| | - Rosario Pasquinelli
- Clinical Oncology Department, Centro de Educación Médica e Investigaciones Clínicas “Norberto Quirno” (CEMIC), Ciudad Autónoma de Buenos Aires, Argentina
| | - Sergio Rivero
- Clinical Oncology Department, Instituto Alexander Fleming, Ciudad Autónoma de Buenos Aires, Argentina
| | - Alexis Ostinelli
- Clinical Oncology Department, Instituto Alexander Fleming, Ciudad Autónoma de Buenos Aires, Argentina
| | - Florencia Perazzo
- Clinical Oncology Department, Centro de Educación Médica e Investigaciones Clínicas “Norberto Quirno” (CEMIC), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
13
|
Chen Q, Xiong J, Ma Y, Wei J, Liu C, Zhao Y. Systemic treatments for breast cancer brain metastasis. Front Oncol 2023; 12:1086821. [PMID: 36686840 PMCID: PMC9853531 DOI: 10.3389/fonc.2022.1086821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/16/2022] [Indexed: 01/07/2023] Open
Abstract
Breast cancer (BC) is the most common cancer in females and BC brain metastasis (BCBM) is considered as the second most frequent brain metastasis. Although the advanced treatment has significantly prolonged the survival in BC patients, the prognosis of BCBM is still poor. The management of BCBM remains challenging. Systemic treatments are important to maintain control of central nervous system disease and improve patients' survival. BCBM medical treatment is a rapidly advancing area of research. With the emergence of new targeted drugs, more options are provided for the treatment of BM. This review features currently available BCBM treatment strategies and outlines novel drugs and ongoing clinical trials that may be available in the future. These treatment strategies are discovered to be more efficacious and potent, and present a paradigm shift in the management of BCBMs.
Collapse
Affiliation(s)
| | | | | | | | - Cuiwei Liu
- *Correspondence: Cuiwei Liu, ; Yanxia Zhao,
| | | |
Collapse
|
14
|
Ferraro E, Seidman AD. Breast Cancer Brain Metastases: Achilles' Heel in Breast Cancer Patients' Care. Cancer Treat Res 2023; 188:283-302. [PMID: 38175350 DOI: 10.1007/978-3-031-33602-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Brain metastases (BM) significantly affect the prognosis as well as the quality of life of breast cancer (BC) patients. Although advancements in neurosurgical and radiotherapy techniques improve local control and symptom management, BM remains associated with a poor prognosis. In addition, the efficacy of currently approved systemic therapies in central nervous system (CNS) compartment is still limited, especially after progression on local therapy. The blood-brain barrier (BBB) has been recognized as a mechanism of primary resistance to many chemotherapeutic agents and targeted therapies due to low drug penetration. Other mechanisms of primary and secondary resistance are still unclear and may vary across the BC subtypes. New small molecules have demonstrated efficacy in BM, in particular for the HER2-positive subtype, with a benefit in survival. A new era has begun in the field of BM, and many trials specifically designed for this population are currently ongoing. The BC research community needs to address this call with the final aim of improving the efficacy of systemic therapy in CNS compartment and ultimately preventing the occurrence of BM.
Collapse
Affiliation(s)
- Emanuela Ferraro
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew D Seidman
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell College of Medicine, New York, NY, USA.
| |
Collapse
|
15
|
Sun H, Xu J, Dai S, Ma Y, Sun T. Breast cancer brain metastasis: Current evidence and future directions. Cancer Med 2023; 12:1007-1024. [PMID: 35822637 PMCID: PMC9883555 DOI: 10.1002/cam4.5021] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the most common cancer in women and the second leading cause of cancer-related deaths after lung cancer. Metastasis of the central nervous system is a terrible event for breast cancer patients, affecting their survival and quality of life. Compared with hormone receptor-positive/human epidermal growth factor receptor 2-negative breast cancer patients, brain metastases are more likely to affect patients with triple-negative breast cancer and human epidermal growth factor receptor 2-positive breast cancer. The treatment of breast cancer has improved greatly in the last two decades. However, brain metastases from breast cancer remain the leading cause of morbidity and mortality. Patients with breast cancer brain metastasis have been in an inferior position due to the lack of clinical research in this field, and they are often explicitly excluded from almost all clinical trials. The occurrence and progression of brain metastases will result in severe cognitive impairment and adverse physical consequences, so we must have a good understanding of the molecular mechanisms of breast cancer brain metastasis. In this article, we have retrieved the latest literature of molecules and pathways associated with breast cancer brain metastasis, summarized common therapy strategies, and discussed the prospects and clinical implications of targeting the molecules involved.
Collapse
Affiliation(s)
- Hongna Sun
- Department of Medical Oncology, Liaoning Cancer Hospital & InstituteCancer Hospital of China Medical UniversityShenyangChina
| | - Junnan Xu
- Department of Medical Oncology, Liaoning Cancer Hospital & InstituteCancer Hospital of China Medical UniversityShenyangChina
| | - Shuang Dai
- Department of Medical Oncology, Lung cancer center, West China HospitalSichuan UniversityChengduChina
| | - Yiwen Ma
- Department of Medical Oncology, Liaoning Cancer Hospital & InstituteCancer Hospital of China Medical UniversityShenyangChina
| | - Tao Sun
- Department of Medical Oncology, Liaoning Cancer Hospital & InstituteCancer Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
16
|
Batista MFFV, Eiriz I, Fitzpatrick A, Le Du F, Braga S, Alpuim Costa D. Refining Therapy in Patients with HER2-Positive Breast Cancer with Central Nervous System Metastasis. Breast Care (Basel) 2022; 17:524-532. [PMID: 36590149 PMCID: PMC9801402 DOI: 10.1159/000526431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 08/07/2022] [Indexed: 01/04/2023] Open
Abstract
Background Brain metastasis (BM) is a major clinical problem in metastatic breast cancer (MBC), occurring in 50% of patients with human epidermal growth factor receptor 2-positive (HER2+) breast cancer. Historically omitted from clinical trials, recent studies of novel HER2-targeted agents have focused on HER2+ BM patients, addressing stable but also progressing BM and leptomeningeal carcinomatosis (LMC). Summary This review aimed to summarize the most relevant data on treating patients with HER2+ BM and LMC. Key Messages The treatment paradigm for patients with HER2+ MBC has changed. Local therapies play an important role, but accumulating evidence on the intracranial activity and clinical benefit of anti-HER2 targeting therapies might lead to a shift in the paradigm on treating BM in the next few years towards more widespread use of systemic therapy.
Collapse
Affiliation(s)
| | - Inês Eiriz
- Oncology Department, Hospital Professor Doutor Fernando Fonseca EPE, Amadora, Portugal
| | - Amanda Fitzpatrick
- Oncology Department, Guy's and St Thomas' Hospital, London, UK
- Oncology Department, The Institute of Cancer Research, London, UK
| | - Fanny Le Du
- Oncology Department, CRLCC Eugène Marquis, Rennes, France
| | - Sofia Braga
- Oncology Department, Hospital Professor Doutor Fernando Fonseca EPE, Amadora, Portugal
- Haematology and Oncology Department, CUF Oncologia, Lisbon, Portugal
| | | |
Collapse
|
17
|
Ma X, Li Y, Li L, Gao C, Liu D, Li H, Zhao Z, Zhao B. Pyrotinib-based treatments in HER2-positive breast cancer patients with brain metastases. Ann Med 2022; 54:3085-3095. [PMID: 36331291 PMCID: PMC9639475 DOI: 10.1080/07853890.2022.2139411] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVES Extensive application of anti-HER2 targeted therapy improves significantly the HER2-positive advanced breast cancer (BC) prognosis, however, it is still difficult to treat brain metastasis. In current study, we explored effective approaches via combining pyrotinib to treat brain metastasis in patients with HER2-positive advanced BC based upon clinical data. MATERIALS AND METHODS Current study included 61 HER2-positive BC patients with brain metastases (BM) who were treated by pyrotinib-based regimens. The systemic regimens included pyrotinib combined with capecitabine, pyrotinib combined with nab-paclitaxel, and pyrotinib combined with vinorelbine. Patients' progression-free survival (PFS), overall survival (OS), clinical benefit rate (CBR) and objective response rate (ORR), as well as drug-related adverse events (AEs) in regard of each combination regimen were analyzed. RESULTS Pyrotinib-based systemic therapy resulted in 8.6 months median PFS (mPFS) and 18.0 months median OS (mOS) among the BM patients. Regarding different regimens, the combination of pyrotinib with nab-paclitaxel was superior to the combination with capecitabine and vinorelbine with respect to PFS and OS. The central nervous system (CNS) ORR did not showcase significant difference among 3 regimens, however, nab-paclitaxel combined regimen obtained the best peripheral ORR (84.6%) (p ≤ .05). CONCLUSIONS Pyrotinib-based combination therapy is safe for HER2-positive brain metastasis treatment. Compared with vinorelbine or capecitabine, pyrotinib combined with nab-paclitaxel is more effective with less toxicity, which is the preferable regimen for HER2-positive brain metastasis.KEY MESSAGESPresent investigation investigated effective methods through combining pyrotinib to treat brain metastasis with HER2-positive advanced brain cancer. The outcomes verified that pyrotinib-based combination therapy was safe and efficient to treat HER2-positive brain metastasis. Therefore, it is effective to treat brain metastasis applying anti-HER2 targeted therapies although pyrotinib showcases efficiency regarding its treatments for the metastasis.
Collapse
Affiliation(s)
- Xiaoping Ma
- Breast Cancer, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Yan Li
- Breast Cancer, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Li Li
- Breast Cancer, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Chunyan Gao
- Breast Cancer, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Dan Liu
- Breast Cancer, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Hongyu Li
- Breast Cancer, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Zhenhui Zhao
- Breast Cancer, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| | - Bing Zhao
- Breast Cancer, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, China
| |
Collapse
|
18
|
Tsuruoka S, Kataoka M, Uwatsu K, Makita K, Tsuruoka K, Takata N, Ishikawa H, Hamamoto Y, Mochizuki T, Kido T. Prognostic value of human epidermal growth factor receptor 2 status and systemic therapy in breast cancer with brain metastases treated with radiotherapy. Asia Pac J Clin Oncol 2022; 19:347-354. [DOI: 10.1111/ajco.13881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 08/25/2022] [Accepted: 09/25/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Shintaro Tsuruoka
- Department of Radiation Oncology National Hospital Organization Shikoku Cancer Center Matsuyama Japan
- Department of Radiology Ehime University Graduate School of Medicine Toon Japan
| | - Masaaki Kataoka
- Department of Radiation Oncology National Hospital Organization Shikoku Cancer Center Matsuyama Japan
- Department of Radiology Saiseikai Imabari Hospital Imabari Japan
| | - Kotaro Uwatsu
- Department of Radiology Ehime University Graduate School of Medicine Toon Japan
| | - Kenji Makita
- Department of Radiology Ehime University Graduate School of Medicine Toon Japan
| | - Kota Tsuruoka
- Department of Radiology Saiseikai Matsuyama Hospital Matsuyama Japan
| | - Noriko Takata
- Department of Radiology Ehime University Graduate School of Medicine Toon Japan
| | - Hirofumi Ishikawa
- Department of Radiology Ehime University Graduate School of Medicine Toon Japan
| | - Yasushi Hamamoto
- Department of Radiation Oncology National Hospital Organization Shikoku Cancer Center Matsuyama Japan
| | - Teruhito Mochizuki
- Department of Radiology Ehime University Graduate School of Medicine Toon Japan
- Department of Radiology I.M. Sechenov First Moscow State Medical University Moscow Russian Federation
| | - Teruhito Kido
- Department of Radiology Ehime University Graduate School of Medicine Toon Japan
| |
Collapse
|
19
|
Tsai C, Nguyen B, Luthra A, Chou JF, Feder L, Tang LH, Strong VE, Molena D, Jones DR, Coit DG, Ilson DH, Ku GY, Cowzer D, Cadley J, Capanu M, Schultz N, Beal K, Moss NS, Janjigian YY, Maron SB. Outcomes and Molecular Features of Brain Metastasis in Gastroesophageal Adenocarcinoma. JAMA Netw Open 2022; 5:e2228083. [PMID: 36001319 PMCID: PMC9403772 DOI: 10.1001/jamanetworkopen.2022.28083] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE Brain metastasis (BrM) in gastroesophageal adenocarcinoma (GEA) is a rare and poorly understood phenomenon associated with poor prognosis. OBJECTIVES To examine the clinical and genomic features of patients with BrM from GEA and evaluate factors associated with survival. DESIGN, SETTING, AND PARTICIPANTS In this single-institution retrospective cohort study, 68 patients with BrM from GEA diagnosed between January 1, 2008, and December 31, 2020, were identified via review of billing codes and imaging reports from the electronic medical record with follow-up through November 3, 2021. Genomic data were derived from the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets clinical sequencing platform. EXPOSURES Treatment with BrM resection and/or radiotherapy. MAIN OUTCOMES AND MEASURES Overall survival after BrM diagnosis. RESULTS Sixty-eight patients (median age at diagnosis, 57.4 years [IQR, 49.8-66.4 years]; 59 [86.8%] male; 55 [85.9%] White) participated in the study. A total of 57 (83.8%) had primary tumors in the distal esophagus or gastroesophageal junction. Median time from initial diagnosis to BrM diagnosis was 16.9 months (IQR, 8.5-27.7 months). Median survival from BrM diagnosis was 8.7 months (95% CI, 5.5-11.5 months). Overall survival was 35% (95% CI, 25%-48%) at 1 year and 24% (95% CI, 16%-37%) at 2 years. In a multivariable analysis, an Eastern Cooperative Oncology Group performance status of 2 or greater (hazard ratio [HR], 4.66; 95% CI, 1.47-14.70; P = .009) and lack of surgical or radiotherapeutic intervention (HR, 7.71; 95% CI, 2.01-29.60; P = .003) were associated with increased risk of all-cause mortality, whereas 3 or more extracranial sites of disease (HR, 1.85; 95% CI, 0.64-5.29; P = .25) and 4 or more BrMs (HR, 2.15; 95% CI, 0.93-4.98; P = .07) were not statistically significant. A total of 31 patients (45.6%) had ERBB2 (formerly HER2 or HER2/neu)-positive tumors, and alterations in ERBB2 were enriched in BrM relative to primary tumors (8 [47.1%] vs 7 [20.6%], P = .05), as were alterations in PTPRT (7 [41.2%] vs 4 [11.8%], P = .03). CONCLUSIONS AND RELEVANCE This study suggests that that a notable proportion of patients with BrM from GEA achieve survival exceeding 1 and 2 years from BrM diagnosis, a more favorable prognosis than previously reported. Good performance status and treatment with combination surgery and radiotherapy were associated with the best outcomes. ERBB2 positivity and amplification as well as PTPRT alterations were enriched in BrM tissue compared with primary tumors; therefore, further study should be pursued to identify whether these variables represent genomic risk factors for BrM development.
Collapse
Affiliation(s)
- Charlton Tsai
- Department of Medicine, New York Presbyterian/Weill Cornell Medicine, New York, New York
| | - Bastien Nguyen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anisha Luthra
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joanne F. Chou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lara Feder
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laura H. Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vivian E. Strong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Molena
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David R. Jones
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniel G. Coit
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David H. Ilson
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Geoffrey Y. Ku
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Darren Cowzer
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John Cadley
- Department of Digital Informatics and Technology Solutions, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nikolaus Schultz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kathryn Beal
- Department of Radiation Oncology and Brain Metastasis Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nelson S. Moss
- Department of Neurosurgery and Brain Metastasis Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yelena Y. Janjigian
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven B. Maron
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
20
|
Mills MN, King W, Soyano A, Pina Y, Czerniecki BJ, Forsyth PA, Soliman H, Han HS, Ahmed KA. Evolving management of HER2+ breast cancer brain metastases and leptomeningeal disease. J Neurooncol 2022; 157:249-269. [PMID: 35244835 DOI: 10.1007/s11060-022-03977-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/23/2022] [Indexed: 12/14/2022]
Abstract
Patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer are at a particularly high risk of breast cancer brain metastasis (BCBM) and leptomeningeal disease (LMD). Improvements in systemic therapy have translated to improved survival for patients with HER2-positive BCBM and LMD. However, the optimal management of these cases is rapidly evolving and requires a multidisciplinary approach. Herein, a team of radiation oncologists, medical oncologists, neuro-oncologists, and breast surgeon created a review of the evolving management of HER2-positive BCBM and LMD. We assess the epidemiology, diagnosis, and evolving treatment options for patients with HER2-positive BCBM and LMD, as well as the ongoing prospective clinical trials enrolling these patients. The management of HER2-positive BCBM and LMD represents an increasingly common challenge that involves the coordination of local and systemic therapy. Advances in systemic therapy have resulted in an improved prognosis, and promising targeted therapies currently under prospective investigation have the potential to further benefit these patients.
Collapse
Affiliation(s)
- Matthew N Mills
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL, 33612, USA
| | - Whitney King
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL, 33612, USA
| | - Aixa Soyano
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Yolanda Pina
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Brian J Czerniecki
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Peter A Forsyth
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Hatem Soliman
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Hyo S Han
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Kamran A Ahmed
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL, 33612, USA.
| |
Collapse
|
21
|
Yu J, Gou W, Shang H, Cui Y, Sun X, Luo L, Hou W, Sun T, Li Y. Design and synthesis of benzodiazepines as brain penetrating PARP-1 inhibitors. J Enzyme Inhib Med Chem 2022; 37:952-972. [PMID: 35317687 PMCID: PMC8942544 DOI: 10.1080/14756366.2022.2053524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The poly (ADP-ribose) polymerase (PARP) inhibitors play a crucial role in cancer therapy. However, most approved PARP inhibitors cannot cross the blood-brain barrier, thus limiting their application in the central nervous system. Here, 55 benzodiazepines were designed and synthesised to screen brain penetrating PARP-1 inhibitors. All target compounds were evaluated for their PARP-1 inhibition activity, and compounds with better activity were selected for further assays in vitro. Among them, compounds H34, H42, H48, and H52 displayed acceptable inhibition effects on breast cancer cells. Also, computational prediction together with the permeability assays in vitro and in vivo proved that the benzodiazepine PARP-1 inhibitors we synthesised were brain permeable. Compound H52 exhibited a B/P ratio of 40 times higher than that of Rucaparib and would be selected to develop its potential use in neurodegenerative diseases. Our study provided potential lead compounds and design strategies for the development of brain penetrating PARP-1 inhibitors.HIGHLIGHTS Structural fusion was used to screen brain penetrating PARP-1 inhibitors. 55 benzodiazepines were evaluated for their PARP-1 inhibition activity. Four compounds displayed acceptable inhibition effects on breast cancer cells. The benzodiazepine PARP-1 inhibitors were proved to be brain permeable.
Collapse
Affiliation(s)
- Jiang Yu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Tianjin, China.,Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Ministry of Education, Shenyang, China
| | - Wenfeng Gou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Tianjin, China
| | - Haihua Shang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Tianjin, China
| | - Yating Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Tianjin, China
| | - Xiao Sun
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Tianjin, China
| | - Lingling Luo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Tianjin, China
| | - Wenbin Hou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Tianjin, China
| | - Tiemin Sun
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Ministry of Education, Shenyang, China
| | - Yiliang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Tianjin, China
| |
Collapse
|
22
|
Ferraro E, Singh J, Patil S, Razavi P, Modi S, Chandarlapaty S, Barrio AV, Malani R, Mellinghoff IK, Boire A, Wen HY, Brogi E, Seidman AD, Norton L, Robson ME, Dang CT. Incidence of brain metastases in patients with early HER2-positive breast cancer receiving neoadjuvant chemotherapy with trastuzumab and pertuzumab. NPJ Breast Cancer 2022; 8:37. [PMID: 35319017 PMCID: PMC8940915 DOI: 10.1038/s41523-022-00380-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 11/01/2021] [Indexed: 11/12/2022] Open
Abstract
The addition of pertuzumab (P) to trastuzumab (H) and neoadjuvant chemotherapy (NAC) has decreased the risk of distant recurrence in early stage HER2-positive breast cancer. The incidence of brain metastases (BM) in patients who achieved pathological complete response (pCR) versus those who do not is unknown. In this study, we sought the incidence of BM in patients receiving HP-containing NAC as well as survival outcome. We reviewed the medical records of 526 early stage HER2-positive patients treated with an HP-based regimen at Memorial Sloan Kettering Cancer Center (MSKCC), between September 1, 2013 to November 1, 2019. The primary endpoint was to estimate the cumulative incidence of BM in pCR versus non-pCR patients; secondary endpoints included disease free-survival (DFS) and overall survival (OS). After a median follow-up of 3.2 years, 7 out of 286 patients with pCR had a BM while 5 out of 240 non-pCR patients had a BM. The 3-year DFS was significantly higher in the pCR group compared to non-pCR group (95% vs 91 %, p = 0.03) and the same trend was observed for overall survival. In our cohort, despite the better survival outcomes of patients who achieved pCR, we did not observe appreciable differences in the incidence of BM by pCR/non-pCR status. This finding suggests that the BM incidence could not be associated with pCR. Future trials with new small molecules able to cross the blood brain barrier should use more specific biomarkers rather than pCR for patients' selection.
Collapse
Affiliation(s)
- Emanuela Ferraro
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jasmeet Singh
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sujata Patil
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Pedram Razavi
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine College, New York, NY, USA
| | - Shanu Modi
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine College, New York, NY, USA
| | - Sarat Chandarlapaty
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine College, New York, NY, USA
| | - Andrea V Barrio
- Breast Cancer Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rachna Malani
- Brain Tumor Center, Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ingo K Mellinghoff
- Brain Tumor Center, Human Oncology and Pathogenesis Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adrienne Boire
- Brain Tumor Center, Human Oncology and Pathogenesis Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hannah Y Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Edi Brogi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew D Seidman
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine College, New York, NY, USA
| | - Larry Norton
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine College, New York, NY, USA
| | - Mark E Robson
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine College, New York, NY, USA
| | - Chau T Dang
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine College, New York, NY, USA.
| |
Collapse
|
23
|
Wu Z, Wan J, Wang J, Meng X, Qian H. Identification of prognostic biomarkers for breast cancer brain metastases based on the bioinformatics analysis. Biochem Biophys Rep 2022; 29:101203. [PMID: 35059509 PMCID: PMC8760394 DOI: 10.1016/j.bbrep.2022.101203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose The prognosis of breast cancer (BC) patients who develop into brain metastases (BMs) is very poor. Thus, it is of great significance to explore the etiology of BMs in BC and identify the key genes involved in this process to improve the survival of BC patients with BMs. Patients and methods The gene expression data and the clinical information of BC patients were downloaded from TCGA and GEO database. Differentially expressed genes (DEGs) in TCGA-BRCA and GSE12276 were overlapped to find differentially expressed metastatic genes (DEMGs). The protein-protein interaction (PPI) network of DEMGs was constructed via STRING database. ClusterProfiler R package was applied to perform the gene ontology (GO) enrichment analysis of DEMGs. The univariate Cox regression analysis and the Kaplan-Meier (K-M) curves were plotted to screen DEMGs associated with the overall survival and the metastatic recurrence survival, which were identified as the key genes associated with the BMs in BC. The immune infiltration and the expressions of immune checkpoints for BC patients with brain relapses and BC patients with other relapses were analyzed respectively. The correlations among the expressions of key genes and the differently infiltrated immune cells or the differentially expressed immune checkpoints were calculated. The gene set enrichment analysis (GSEA) of each key gene was conducted to investigate the potential mechanisms of key genes involved in BC patients with BMs. Moreover, CTD database was used to predict the drug-gene interaction network of key genes. Results A total of 154 DEGs were identified in BC patients at M0 and M1 in TCGA database. A total of 667 DEGs were identified in BC patients with brain relapses and with other relapses. By overlapping these DEGs, 17 DEMGs were identified, which were enriched in the cell proliferation related biological processes and the immune related molecular functions. The univariate Cox regression analysis and the Kaplan-Meier curves revealed that CXCL9 and GPR171 were closely associated with the overall survival and the metastatic recurrence survival and were identified as key genes associated with BMs in BC. The analyses of immune infiltration and immune checkpoint expressions showed that there was a significant difference of the immune microenvironment between brain relapses and other relapses in BC. GSEA indicated that CXCL9 and GPR171 may regulate BMs in BC via the immune-related pathways. Conclusion Our study identified the key genes associated with BMs in BC patients and explore the underlying mechanisms involved in the etiology of BMs in BC. These findings may provide a promising approach for the treatments of BC patients with BMs. CXCL9 and GPR171, as the key genes, were closely associated with the prognosis of brain metastases in breast cancer. There was a significant difference of the immune microenvironment between brain and other metastases in breast cancer. We revealed candidate drugs which associated with the key genes of breast cancer patients with brain metastases. A series of bioinformatic analysis methods were used in this article.
Collapse
Affiliation(s)
| | - Jinghai Wan
- Corresponding author. National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Neurosurgery, Panjiayuan 17#, Chaoyang District, Beijing, China.
| | | | | | | |
Collapse
|
24
|
Carausu M, Carton M, Cabel L, Patsouris A, Levy C, Verret B, Pasquier D, Debled M, Gonçalves A, Desmoulins I, Lecouillard I, Bachelot T, Ferrero JM, Eymard JC, Mouret-Reynier MA, Chevrot M, De Maio E, Uwer L, Frenel JS, Leheurteur M, Petit T, Darlix A, Bozec L. Clinicopathological characteristics and prognosis of breast cancer patients with isolated central nervous system metastases in the multicentre ESME database. Ther Adv Med Oncol 2022; 14:17588359221077082. [PMID: 35237352 PMCID: PMC8883300 DOI: 10.1177/17588359221077082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 01/12/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND As a result of progress in diagnosis and treatment, there is a growing prevalence of metastatic breast cancer (MBC) with isolated CNS metastases. This study describes the largest-to-date real-life cohort of this clinical setting and compares it to other clinical presentations. METHODS We retrospectively analysed the French Epidemiological Strategy and Medical Economics (ESME) MBC database including patients who initiated treatment for MBC between 2008 and 2016. Progression-free survival (PFS) and overall survival (OS) were estimated using the Kaplan-Meier method. Descriptive statistics and multivariate Cox model were used. RESULTS Of 22,266 patients, 647 (2.9%) and 929 (4.2%) patients had isolated first-site CNS metastases or combined with extra-CNS metastases, with longer OS for the group with isolated CNS metastases (16.9 versus 13.9 months, adjusted HR = 1.69 (95% CI: 1.50-1.91), p < 0.001). Among the 541 (2.4%) patients with isolated CNS metastases and no intrathecal therapy (excluding leptomeningeal metastases), HER2+ cases were preponderant over TN or HR+ /HER2- cases (41.6% versus 26.1% versus 28.5%, respectively, p < 0.01). The treatment strategy consisted of a combination of local treatment and systemic therapy (49.2%), local treatment only (35.5%) or systemic therapy only (11.4%), or symptomatic therapy only (3.9%). Median PFS was 6.1 months (95% CI: 5.7-6.8). Median OS was 20.7 months (95% CI: 17.3-24.3), reaching 37.9 months (95% CI: 25.9-47.6) in the HR+ /HER2+ subgroup. Older age, TN subtype, MBC-free interval of 6-12 months, lower performance status, and WBRT were associated with poorer survival. Patients who received systemic therapy within 3 months from MBC diagnosis had longer OS (24.1 versus 16.1 months, p = 0.031), but this was not significant on multivariate analysis [HR = 1.0 (95% CI: 0.7-1.3), p = 0.806]. CONCLUSIONS Patients with isolated CNS metastases at MBC diagnosis represent a distinct population for which the role of systemic therapy needs to be further investigated in prospective studies.
Collapse
Affiliation(s)
- Marcela Carausu
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
| | - Matthieu Carton
- Department of Biostatistics, Institut Curie, Saint-Cloud, France
| | - Luc Cabel
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
| | - Anne Patsouris
- Department of Medical Oncology, Institut de Cancérologie de l’Ouest, Angers, France
| | - Christelle Levy
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Benjamin Verret
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France
| | - David Pasquier
- Department of Radiation Oncology, Centre Oscar Lambret, CRIStAL UMR CNRS 9189, Lille University, Lille, France
| | - Marc Debled
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Isabelle Desmoulins
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | | | - Thomas Bachelot
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Jean-Marc Ferrero
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | | | | | | | - Eleonora De Maio
- Department of Medical Oncology, Institut Claudius Regaud – IUCT Oncopole, Toulouse, France
| | - Lionel Uwer
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandœuvre-lès-Nancy, France
| | - Jean-Sébastien Frenel
- Department of Medical Oncology, Institut de Cancérologie de l’Ouest–René Gauducheau, Saint-Herblain, France
| | | | - Thierry Petit
- Department of Medical Oncology, Centre Paul Strauss, Strasbourg, France
| | - Amélie Darlix
- Department of Medical Oncology, Institut régional du Cancer de Montpellier (ICM), Institut de Génomique Fonctionnelle, INSERM U1191-CNRS UMR 5203, Université de Montpellier, Montpellier, France
| | - Laurence Bozec
- Department of Medical Oncology, Institut Curie, 35 rue Dailly, 92210 Saint-Cloud, France
| |
Collapse
|
25
|
Lim M, Nguyen TH, Niland C, Reid LE, Jat PS, Saunus JM, Lakhani SR. Landscape of Epidermal Growth Factor Receptor Heterodimers in Brain Metastases. Cancers (Basel) 2022; 14:cancers14030533. [PMID: 35158800 PMCID: PMC8833370 DOI: 10.3390/cancers14030533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary HER2+ breast cancer patients are treated with agents that tag HER2+ tumour cells for elimination by the immune system, down-modulate HER2 activity and/or block the formation of HER2 dimers, including the neuregulin-1 receptor, HER2-HER3. HER2-targeted therapies prolong survival by lowering the risk of relapse, but do not prevent brain metastases. The reasons for this are not fully understood. We quantified HER2-HER3 dimers in 203 brain metastases, and 34 primary breast tumour samples. Dimer frequency was relatively high in brain metastases from breast, ovarian, lung and kidney cancers, and in brain metastases compared to patient-matched breast tumours; but did not reliably correlate with HER2/HER3 expression or activation. In in vitro experiments, pertuzumab failed to suppress HER2-HER3 dimers in HER2+ breast cancer cells provided with a saturating concentration of neuregulin-1. These findings may provide insights about the differences in intracranial versus extracranial efficacy of HER2-targeted therapies. Abstract HER2+ breast cancer patients have an elevated risk of developing brain metastases (BM), despite adjuvant HER2-targeted therapy. The mechanisms underpinning this reduced intracranial efficacy are unclear. We optimised the in situ proximity ligation assay (PLA) for detection of the high-affinity neuregulin-1 receptor, HER2-HER3 (a key target of pertuzumab), in archival tissue samples and developed a pipeline for high throughput extraction of PLA data from fluorescent microscope image files. Applying this to a large BM sample cohort (n = 159) showed that BM from breast, ovarian, lung and kidney cancers have higher HER2-HER3 levels than other primary tumour types (melanoma, colorectal and prostate cancers). HER2 status, and tumour cell membrane expression of pHER2(Y1221/1222) and pHER3(Y1222) were positively, but not exclusively, associated with HER2-HER3 frequency. In an independent cohort (n = 78), BM had significantly higher HER2-HER3 levels than matching primary tumours (p = 0.0002). For patients who had two craniotomy procedures, HER2-HER3 dimer levels were lower in the consecutive lesion (n = 7; p = 0.006). We also investigated the effects of trastuzumab and pertuzumab on five different heterodimers in vitro: HER2-EGFR, HER2-HER4, HER2-HER3, HER3-HER4, HER3-EGFR. Treatment significantly altered the absolute frequencies of individual complexes in SKBr3 and/or MDA-MB-361 cells, but in the presence of neuregulin-1, the overall distribution was not markedly altered, with HER2-HER3 and HER2-HER4 remaining predominant. Together, these findings suggest that markers of HER2 and HER3 expression are not always indicative of dimerization, and that pertuzumab may be less effective at reducing HER2-HER3 dimerization in the context of excess neuregulin.
Collapse
Affiliation(s)
- Malcolm Lim
- Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Herston, QLD 4029, Australia; (M.L.); (C.N.); (L.E.R.)
| | - Tam H. Nguyen
- Flow Cytometry and Imaging Facility, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia;
| | - Colleen Niland
- Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Herston, QLD 4029, Australia; (M.L.); (C.N.); (L.E.R.)
| | - Lynne E. Reid
- Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Herston, QLD 4029, Australia; (M.L.); (C.N.); (L.E.R.)
| | - Parmjit S. Jat
- Department of Neurodegenerative Disease and MRC Prion Unit, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK;
| | - Jodi M. Saunus
- Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Herston, QLD 4029, Australia; (M.L.); (C.N.); (L.E.R.)
- Correspondence: (J.M.S.); (S.R.L.)
| | - Sunil R. Lakhani
- Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Herston, QLD 4029, Australia; (M.L.); (C.N.); (L.E.R.)
- Pathology Queensland, Royal Brisbane Women’s Hospital, Herston, QLD 4029, Australia
- Correspondence: (J.M.S.); (S.R.L.)
| |
Collapse
|
26
|
Parida PK, Marquez-Palencia M, Nair V, Kaushik AK, Kim K, Sudderth J, Quesada-Diaz E, Cajigas A, Vemireddy V, Gonzalez-Ericsson PI, Sanders ME, Mobley BC, Huffman K, Sahoo S, Alluri P, Lewis C, Peng Y, Bachoo RM, Arteaga CL, Hanker AB, DeBerardinis RJ, Malladi S. Metabolic diversity within breast cancer brain-tropic cells determines metastatic fitness. Cell Metab 2022; 34:90-105.e7. [PMID: 34986341 PMCID: PMC9307073 DOI: 10.1016/j.cmet.2021.12.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/10/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
HER2+ breast cancer patients are presented with either synchronous (S-BM), latent (Lat), or metachronous (M-BM) brain metastases. However, the basis for disparate metastatic fitness among disseminated tumor cells of similar oncotype within a distal organ remains unknown. Here, employing brain metastatic models, we show that metabolic diversity and plasticity within brain-tropic cells determine metastatic fitness. Lactate secreted by aggressive metastatic cells or lactate supplementation to mice bearing Lat cells limits innate immunosurveillance and triggers overt metastasis. Attenuating lactate metabolism in S-BM impedes metastasis, while M-BM adapt and survive as residual disease. In contrast to S-BM, Lat and M-BM survive in equilibrium with innate immunosurveillance, oxidize glutamine, and maintain cellular redox homeostasis through the anionic amino acid transporter xCT. Moreover, xCT expression is significantly higher in matched M-BM brain metastatic samples compared to primary tumors from HER2+ breast cancer patients. Inhibiting xCT function attenuates residual disease and recurrence in these preclinical models.
Collapse
Affiliation(s)
- Pravat Kumar Parida
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mauricio Marquez-Palencia
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vidhya Nair
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Akash K Kaushik
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kangsan Kim
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jessica Sudderth
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eduardo Quesada-Diaz
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ambar Cajigas
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vamsidhara Vemireddy
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Paula I Gonzalez-Ericsson
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | - Melinda E Sanders
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | - Bret C Mobley
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | - Kenneth Huffman
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sunati Sahoo
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Prasanna Alluri
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Cheryl Lewis
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yan Peng
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Robert M Bachoo
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Carlos L Arteaga
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ariella B Hanker
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ralph J DeBerardinis
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Srinivas Malladi
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
27
|
Combination of tucatinib and neural stem cells secreting anti-HER2 antibody prolongs survival of mice with metastatic brain cancer. Proc Natl Acad Sci U S A 2022; 119:2112491119. [PMID: 34969858 PMCID: PMC8740706 DOI: 10.1073/pnas.2112491119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2021] [Indexed: 12/27/2022] Open
Abstract
Brain metastases are among the most severe complications of systemic breast cancer, and overexpression of the human epidermal growth factor receptor 2 (HER2) in breast cancer cells increases the incidence of brain metastases in patients. In this study, we engineered the human-derived, tumor cell tropic neural stem cells LM-NSC008 (LM008) to continuously secrete antibodies against HER2. These anti-HER2 antibodies impaired tumor cell proliferation by inhibiting the PI3K-Akt signaling pathway in HER2+ breast cancer cells in vitro. Importantly, our results demonstrate that the therapeutic combinatorial regimen consisting of LM-NSC008 anti-HER2 antibody-secreting cells and the HER2 kinase inhibitor tucatinib provide therapeutic benefit and prolong survival in preclinical models of HER2+ breast cancer brain metastases. Brain metastases are a leading cause of death in patients with breast cancer. The lack of clinical trials and the presence of the blood–brain barrier limit therapeutic options. Furthermore, overexpression of the human epidermal growth factor receptor 2 (HER2) increases the incidence of breast cancer brain metastases (BCBM). HER2-targeting agents, such as the monoclonal antibodies trastuzumab and pertuzumab, improved outcomes in patients with breast cancer and extracranial metastases. However, continued BCBM progression in breast cancer patients highlighted the need for novel and effective targeted therapies against intracranial metastases. In this study, we engineered the highly migratory and brain tumor tropic human neural stem cells (NSCs) LM008 to continuously secrete high amounts of functional, stable, full-length antibodies against HER2 (anti-HER2Ab) without compromising the stemness of LM008 cells. The secreted anti-HER2Ab impaired tumor cell proliferation in vitro in HER2+ BCBM cells by inhibiting the PI3K-Akt signaling pathway and resulted in a significant benefit when injected in intracranial xenograft models. In addition, dual HER2 blockade using anti-HER2Ab LM008 NSCs and the tyrosine kinase inhibitor tucatinib significantly improved the survival of mice in a clinically relevant model of multiple HER2+ BCBM. These findings provide compelling evidence for the use of HER2Ab-secreting LM008 NSCs in combination with tucatinib as a promising therapeutic regimen for patients with HER2+ BCBM.
Collapse
|
28
|
Pituitary metastasis in HER-2 positive breast cancer with initial presentation of hypernatremia: Two case reports. CURRENT PROBLEMS IN CANCER: CASE REPORTS 2021. [DOI: 10.1016/j.cpccr.2021.100107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
29
|
Carney CP, Pandey N, Kapur A, Woodworth GF, Winkles JA, Kim AJ. Harnessing nanomedicine for enhanced immunotherapy for breast cancer brain metastases. Drug Deliv Transl Res 2021; 11:2344-2370. [PMID: 34716900 PMCID: PMC8568876 DOI: 10.1007/s13346-021-01039-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2021] [Indexed: 12/15/2022]
Abstract
Brain metastases (BMs) are the most common type of brain tumor, and the incidence among breast cancer (BC) patients has been steadily increasing over the past two decades. Indeed, ~ 30% of all patients with metastatic BC will develop BMs, and due to few effective treatments, many will succumb to the disease within a year. Historically, patients with BMs have been largely excluded from clinical trials investigating systemic therapies including immunotherapies (ITs) due to limited brain penetration of systemically administered drugs combined with previous assumptions that BMs are poorly immunogenic. It is now understood that the central nervous system (CNS) is an immunologically distinct site and there is increasing evidence that enhancing immune responses to BCBMs will improve patient outcomes and the efficacy of current treatment regimens. Progress in IT for BCBMs, however, has been slow due to several intrinsic limitations to drug delivery within the brain, substantial safety concerns, and few known targets for BCBM IT. Emerging studies demonstrate that nanomedicine may be a powerful approach to overcome such limitations, and has the potential to greatly improve IT strategies for BMs specifically. This review summarizes the evidence for IT as an effective strategy for BCBM treatment and focuses on the nanotherapeutic strategies currently being explored for BCBMs including targeting the blood-brain/tumor barrier (BBB/BTB), tumor cells, and tumor-supporting immune cells for concentrated drug release within BCBMs, as well as use of nanoparticles (NPs) for delivering immunomodulatory agents, for inducing immunogenic cell death, or for potentiating anti-tumor T cell responses.
Collapse
Affiliation(s)
- Christine P Carney
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Nikhil Pandey
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Anshika Kapur
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jeffrey A Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery and Neurosurgery, University of Maryland School of Medicine, 800 West Baltimore St., Baltimore, MD, 21201, USA.
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA.
- Departments of Neurosurgery, Pharmacology, and Pharmaceutical Sciences, University of Maryland School of Medicine, 655 W Baltimore St., Baltimore, MD, 21201, USA.
| |
Collapse
|
30
|
Oldenburger E, Cappelle S, Wildiers H, Punie K, Daisne JF, Lambrecht M. Solitary pituitary metastasis from HER2-positive breast cancer treated with stereotactic radiotherapy – a case report. CURRENT PROBLEMS IN CANCER: CASE REPORTS 2021. [DOI: 10.1016/j.cpccr.2021.100081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
31
|
Li C, Bian X, Liu Z, Wang X, Song X, Zhao W, Liu Y, Yu Z. Effectiveness and safety of pyrotinib-based therapy in patients with HER2-positive metastatic breast cancer: A real-world retrospective study. Cancer Med 2021; 10:8352-8364. [PMID: 34672424 PMCID: PMC8633258 DOI: 10.1002/cam4.4335] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/22/2021] [Accepted: 09/20/2021] [Indexed: 11/07/2022] Open
Abstract
The previous studies had demonstrated the promising effectiveness and acceptable safety of pyrotinib in patients with HER2‐positive metastatic breast cancer. We aimed to investigate the real‐world data of pyrotinib in complex clinical practice and complement the findings of clinical trials. Two hundred and eighteen patients were included for effectiveness analysis. A total of 62.0% had received two or more lines of systematic therapy, and 95.4% had been exposed to prior anti‐HER2 therapy, with 95.4% receiving trastuzumab, 5.0% receiving pertuzumab, and 40.8% receiving lapatinib. The median progression‐free survival (PFS) was 9.3 months and the objective response rate (ORR) was 44.0%. Patients treated with pyrotinib‐based therapy as first, second, or later line had a median PFS of 15.0, 10.3, and 6.8 months, respectively. Patients treated with pyrotinib and trastuzumab received significant benefit in terms of median PFS compared with pyrotinib alone (10.7 (9.1–12.3) vs. 8.8 (8.1–9.5), p = 0.016). Patients pretreated with lapatinib had a median PFS of 6.9 months. The median PFS time was 7.0 months in patients with brain metastasis. Multivariate Cox regression analyses showed that lines of pyrotinib‐based therapy (1 vs. 2 vs. ≥3), prior treatment with lapatinib, and combination treatments with trastuzumab proved to be independent predictors of PFS. Two hundred and forty‐eight patients were included in the safety analysis, and the results showed that the toxicity of pyrotinib was tolerable, with the most common grade 3/4 adverse event being diarrhea (19.8%). Pyrotinib‐based therapy demonstrated promising efficacy and tolerable toxicity in first‐, second‐, and later‐line treatments and in lapatinib‐treated patients. The combination of pyrotinib and trastuzumab showed advantages in PFS, even for patients resisting trastuzumab. Pyrotinib‐based therapy could be the preferred choice for brain metastasis patients, especially when combined with brain radiotherapy.
Collapse
Affiliation(s)
- Chao Li
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaoli Bian
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Breast Surgery, Heze Municipal Hospital, Heze, China
| | - Zhaoyun Liu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xinzhao Wang
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiang Song
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Zhao
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yansong Liu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhiyong Yu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
32
|
Sun Y, Chen B, Li J, Peng L, Li S, Yu X, Li L. Real-World Analysis of the Efficacy and Safety of a Novel Irreversible HER2 Tyrosine Kinase Inhibitor Pyrotinib in Patients with HER2-Positive Metastatic Breast Cancer. Cancer Manag Res 2021; 13:7165-7174. [PMID: 34548820 PMCID: PMC8449550 DOI: 10.2147/cmar.s321428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/19/2021] [Indexed: 11/23/2022] Open
Abstract
Background As a novel irreversible pan-ErbB inhibitor recently approved in China, pyrotinib has exhibited promising anticancer efficacy and acceptable safety profile in HER2-positive metastatic breast cancer (mBC). The aim of this retrospective study was to estimate the efficacy and safety of pyrotinib treatment in Chinese mBC patients. Methods We retrospectively reviewed the real-world clinicopathological and treatment data of HER2-positive mBC patients receiving pyrotinib-based treatment from August 2018 to July 2019 in Qilu Hospital of Shandong University and other medical centers of Shandong Province in China. Results A total of 64 patients treated with pyrotinib were included for analysis, and the median follow-up duration was 260 days (interquartile range, 199.0 to 339.0 days). Fifty-nine (92.2%) patients had been previously treated with trastuzumab and/or T-DM1, while 11 (17.2%) patients had been exposed to lapatinib. The objective response rate (ORR) of all patients was 73.4%, and the disease control rate (DCR) was 98.4%, with a clinical benefit rate (CBR) of 87.5%. Patients with exposure to lapatinib responded well to pyrotinib-based treatment, although the ORR was significantly lower compared with that of patients without exposure to lapatinib (44.1% vs 77.5%, p=0.037). Previous lapatinib exposure was negatively associated with the objective response of pyrotinib treatment (odds ratio [OR]=0.248, 95% confidence interval [CI] 0.063–0.970, p=0.045). The median progression-free survival (mPFS) for patients with previous lapatinib exposure and patients with visceral metastasis was 299 days (95% CI 240.1–357.9 days) and 359 days (95% CI 258.3–459.7 days), respectively. But the mPFS of the whole cohort has not been reached until the cut-off date. Cox multivariate analysis revealed that only visceral metastasis was an independent predictor of significantly shorter PFS (p=0.041) but not previous exposure to lapatinib (p=0.092). Diarrhea (28.1%), hand-foot syndrome (17.2%), and neutropenia (9.4%) were the most common grade 3 adverse events associated with pyrotinib treatment. Conclusion Pyrotinib is highly beneficial to HER2-positive metastatic breast cancer patients, even in patients with previous lapatinib exposure. Pyrotinib is a feasible replacement of lapatinib in combination with chemotherapeutic drugs or as a monotherapy. Adverse effects are tolerable and easily manageable.
Collapse
Affiliation(s)
- Ying Sun
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Pathology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, 250012, People's Republic of China.,Department of Medical Oncology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, Shandong Province, People's Republic of China
| | - Beibei Chen
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, People's Republic of China.,Department of Radiation Oncology, Heze Municipal Hospital, Heze, 274031, Shandong Province, People's Republic of China
| | - Jisheng Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, People's Republic of China
| | - Ling Peng
- Department of Respiratory Disease, Zhejiang Provincial People's Hospital, Hangzhou, 310000, Zhejiang Province, People's Republic of China
| | - Shuguang Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, People's Republic of China
| | - Xuejun Yu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, People's Republic of China
| | - Li Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, People's Republic of China
| |
Collapse
|
33
|
Garcia-Alvarez A, Papakonstantinou A, Oliveira M. Brain Metastases in HER2-Positive Breast Cancer: Current and Novel Treatment Strategies. Cancers (Basel) 2021; 13:2927. [PMID: 34208287 PMCID: PMC8230933 DOI: 10.3390/cancers13122927] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 01/08/2023] Open
Abstract
Development of brain metastases can occur in up to 30-50% of patients with breast cancer, representing a significant impact on an individual patient in terms of survival and quality of life. Patients with HER2-positive breast cancer have an increased risk of developing brain metastases; however, screening for brain metastases is not currently recommended due to the lack of robust evidence to support survival benefit. In recent years, several novel anti-HER2 agents have led to significant improvements in the outcomes of HER2-positive metastatic breast cancer. Despite these advances, brain and leptomeningeal metastases from HER2-positive breast cancer remain a significant cause of morbidity and mortality, and their optimal management remains an unmet need. This review presents an update on the current and novel treatment strategies for patients with brain metastases from HER2-positive breast cancer and discusses the open questions in the field.
Collapse
Affiliation(s)
| | - Andri Papakonstantinou
- Breast Cancer Group, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain;
- Department of Oncology-Pathology, Karolinska Institute, 17177 Stockholm, Sweden
- Department of Breast Cancer, Endocrine Tumors and Sarcoma, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Mafalda Oliveira
- Medical Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain;
- Breast Cancer Group, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain;
| |
Collapse
|
34
|
Pellerino A, Brastianos PK, Rudà R, Soffietti R. Leptomeningeal Metastases from Solid Tumors: Recent Advances in Diagnosis and Molecular Approaches. Cancers (Basel) 2021; 13:2888. [PMID: 34207653 PMCID: PMC8227730 DOI: 10.3390/cancers13122888] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 06/05/2021] [Indexed: 12/13/2022] Open
Abstract
Leptomeningeal metastases (LM) from solid tumors represent an unmet need of increasing importance due to an early use of MRI for diagnosis and improvement of outcome of some molecular subgroups following targeted agents and immunotherapy. In this review, we first discussed factors limiting the efficacy of targeted agents in LM, such as the molecular divergence between primary tumors and CNS lesions and CNS barriers at the level of the normal brain, brain tumors and CSF. Further, we reviewed pathogenesis and experimental models and modalities, such as MRI (with RANO and ESO/ESMO criteria), CSF cytology and liquid biopsy, to improve diagnosis and monitoring following therapy. Efficacy and limitations of targeted therapies for LM from EGFR-mutant and ALK-rearranged NSCLC, HER2-positive breast cancer and BRAF-mutated melanomas are reported, including the use of intrathecal administration or modification of traditional cytotoxic compounds. The efficacy of checkpoint inhibitors in LM from non-druggable tumors, in particular triple-negative breast cancer, is discussed. Last, we focused on some recent techniques to improve drug delivery.
Collapse
Affiliation(s)
- Alessia Pellerino
- Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; (R.R.); (R.S.)
| | - Priscilla K. Brastianos
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02115, USA;
| | - Roberta Rudà
- Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; (R.R.); (R.S.)
- Department of Neurology, Castelfranco Veneto and Brain Tumor Board Treviso Hospital, 31100 Treviso, Italy
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; (R.R.); (R.S.)
| |
Collapse
|
35
|
Ulrich L, Okines AFC. Treating Advanced Unresectable or Metastatic HER2-Positive Breast Cancer: A Spotlight on Tucatinib. BREAST CANCER (DOVE MEDICAL PRESS) 2021; 13:361-381. [PMID: 34079368 PMCID: PMC8164963 DOI: 10.2147/bctt.s268451] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/24/2021] [Indexed: 12/17/2022]
Abstract
The management of HER2 positive breast cancer has been transformed by the development of targeted therapies. Dual blockade with the monoclonal antibodies, trastuzumab and pertuzumab, added to first-line taxane chemotherapy and second-line therapy with the antibody-drug conjugate, T-DM1, are internationally agreed standards of care for advanced HER2 positive breast cancer, where available. However, until recently, options for patients for third-line therapy and beyond were of modest efficacy or limited by toxicity. In 2019, the results of trials of two exciting new agents for this space were presented. A third-generation HER2 tyrosine kinase inhibitor, tucatinib, combines the efficacy of the second-generation drug, neratinib, with a more manageable toxicity profile and has become a new standard of care after T-DM1, in combination with capecitabine and trastuzumab. The antibody-drug conjugate, trastuzumab deruxtecan, demonstrated remarkable efficacy in heavily pre-treated patients and received accelerated approval in the United States, whilst confirmatory Phase 3 trials are completed. This review will discuss the available data for the post-T-DM1 setting, focusing on tyrosine kinase inhibitors including tucatinib.
Collapse
Affiliation(s)
- Lara Ulrich
- Department of Breast Oncology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Alicia F C Okines
- Department of Breast Oncology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
36
|
Erickson AW, Habbous S, Hoey C, Jerzak KJ, Das S. Dual- versus single-agent HER2 inhibition and incidence of intracranial metastatic disease: a systematic review and meta-analysis. NPJ Breast Cancer 2021; 7:17. [PMID: 33602948 PMCID: PMC7892568 DOI: 10.1038/s41523-021-00220-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/07/2021] [Indexed: 12/19/2022] Open
Abstract
Observational studies have suggested that HER2 inhibition with trastuzumab may be associated with an increased incidence of intracranial metastatic disease (IMD) due to its ability to prolong survival. We hypothesized that prolonged survival associated with dual-agent HER2 inhibition may be associated with an even higher incidence of IMD. This study pooled estimates of IMD incidence and survival among patients with HER2-positive breast cancer receiving dual- versus single-agent HER2 targeted therapy, as well as trastuzumab versus chemotherapy, observation, or another HER2-targeted agent. We searched PubMed, EMBASE, and CENTRAL from inception to 25 March 2020. We included randomized controlled trials that reported IMD incidence for patients with HER2-positive breast cancer receiving trastuzumab as the experimental or control arm irrespective of disease stage. Among 465 records identified, 19 randomized controlled trials (32,572 patients) were included. Meta-analysis of four studies showed that dual HER2-targeted therapy was associated with improved overall survival (HR 0.76; 95% CI, 0.66–0.87) and progression-free survival (HR 0.77; 95% CI, 0.68–0.87) compared to single HER2-targeted therapy, but the risk of IMD was similar (RR 1.03; 95% CI, 0.83–1.27). Our study challenges the hypothesis that prolonged survival afforded by improved extracranial disease control is associated with increased IMD incidence.
Collapse
Affiliation(s)
| | - Steven Habbous
- Ontario Health (Cancer Care Ontario), Toronto, ON, Canada
| | - Christianne Hoey
- Evaluative Clinical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Katarzyna J Jerzak
- Department of Medicine, Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Canada
| | - Sunit Das
- Institute of Medical Science, University of Toronto, Toronto, Canada. .,Division of Neurosurgery, University of Toronto, Toronto, Canada. .,Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, Canada.
| |
Collapse
|
37
|
Al-Akhrass H, Conway JRW, Poulsen ASA, Paatero I, Kaivola J, Padzik A, Andersen OM, Ivaska J. A feed-forward loop between SorLA and HER3 determines heregulin response and neratinib resistance. Oncogene 2021; 40:1300-1317. [PMID: 33420373 PMCID: PMC7892347 DOI: 10.1038/s41388-020-01604-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/23/2020] [Accepted: 12/03/2020] [Indexed: 01/29/2023]
Abstract
Current evidence indicates that resistance to the tyrosine kinase-type cell surface receptor (HER2)-targeted therapies is frequently associated with HER3 and active signaling via HER2-HER3 dimers, particularly in the context of breast cancer. Thus, understanding the response to HER2-HER3 signaling and the regulation of the dimer is essential to decipher therapy relapse mechanisms. Here, we investigate a bidirectional relationship between HER2-HER3 signaling and a type-1 transmembrane sorting receptor, sortilin-related receptor (SorLA; SORL1). We demonstrate that heregulin-mediated signaling supports SorLA transcription downstream of the mitogen-activated protein kinase pathway. In addition, we demonstrate that SorLA interacts directly with HER3, forming a trimeric complex with HER2 and HER3 to attenuate lysosomal degradation of the dimer in a Ras-related protein Rab4-dependent manner. In line with a role for SorLA in supporting the stability of the HER2 and HER3 receptors, loss of SorLA compromised heregulin-induced cell proliferation and sensitized metastatic anti-HER2 therapy-resistant breast cancer cells to neratinib in cancer spheroids in vitro and in vivo in a zebrafish brain xenograft model.
Collapse
Affiliation(s)
- Hussein Al-Akhrass
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland.
| | - James R W Conway
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Annemarie Svane Aavild Poulsen
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of biomedicine, Aarhus University, Aarhus, Denmark
| | - Ilkka Paatero
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Jasmin Kaivola
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Artur Padzik
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Olav M Andersen
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of biomedicine, Aarhus University, Aarhus, Denmark
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland.
| |
Collapse
|
38
|
Masmudi-Martín M, Zhu L, Sanchez-Navarro M, Priego N, Casanova-Acebes M, Ruiz-Rodado V, Giralt E, Valiente M. Brain metastasis models: What should we aim to achieve better treatments? Adv Drug Deliv Rev 2021; 169:79-99. [PMID: 33321154 DOI: 10.1016/j.addr.2020.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/16/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023]
Abstract
Brain metastasis is emerging as a unique entity in oncology based on its particular biology and, consequently, the pharmacological approaches that should be considered. We discuss the current state of modelling this specific progression of cancer and how these experimental models have been used to test multiple pharmacologic strategies over the years. In spite of pre-clinical evidences demonstrating brain metastasis vulnerabilities, many clinical trials have excluded patients with brain metastasis. Fortunately, this trend is getting to an end given the increasing importance of secondary brain tumors in the clinic and a better knowledge of the underlying biology. We discuss emerging trends and unsolved issues that will shape how we will study experimental brain metastasis in the years to come.
Collapse
|
39
|
Wei H, Zhang YJ, Yu T, Yan XY, Jiang Q. Methodological quality evaluation of systematic reviews or meta-analysis of trastuzumab-based therapy for breast cancer: A systematic review. Medicine (Baltimore) 2021; 100:e24389. [PMID: 33530234 PMCID: PMC7850684 DOI: 10.1097/md.0000000000024389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/21/2020] [Accepted: 12/29/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND To evaluate the methodological quality of systematic reviews (SRs) or meta-analysis of trastuzumab-based therapy for breast cancer. METHODS We searched the PubMed, EMBASE, Web of science, Cochrane library, international prospective register of systematic reviews, Chinese BioMedical Literature Database, Wan Fang, China National Knowledge Infrastructure and VIP database for SRs or meta-analysis. The methodological quality of included literatures was appraised by risk of bias in systematic review (ROBIS) tool. RESULTS Twenty three eligible systematic reviews or meta-analysis were included. Only 2 systematic reviews provided protocol. The most frequently searched databases were PubMed, MEDLINE, EMBASE, and the Cochrane. The two-reviewers model described in the screening for eligible original articles, data extraction, and methodological quality evaluation had 30%, 61%, and 26%, respectively. In methodological quality assessment, 52% SRs or meta-analysis used the Jadad scoring or Cochrane reviewer' handbook. Research question were well matched to all SRs or meta-analysis in phase 1 and 35% of them evaluated "high" risk bias in study eligibility criteria. The "high" risk of bias in all non-Cochrane SRs or meta-analyses, which involve methods used to identify and/or select studies. And more than half SRs or meta-analysis had a high risk of bias in data collection and study appraisal. More than two-third of SRs or meta-analysis were accomplished with high risk of bias in the synthesis and findings. CONCLUSIONS The study indicated poor methodological and reporting quality of SRs/meta-analysis assessing trastuzumab-based therapy for breast cancer. Registration or publishing the protocol and the reporting followed the PRISMA checklist are recommended in future research.
Collapse
Affiliation(s)
- Hua Wei
- School of Pharmacy, Chengdu Medical College, Chengdu
- Department of Pharmacy
| | - Yong-Jun Zhang
- Department of Anesthesiology, Dujiangyan People's Hospital, Dujiangyan Medical Center, Dujiangyan
| | - Ting Yu
- Department of Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Centre, School of Medicine, University of Electronic Science and Technology of China
| | - Xiao-Yan Yan
- School of Pharmacy, Chengdu Medical College, Chengdu
| | - Qian Jiang
- Department of Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Centre, School of Medicine, University of Electronic Science and Technology of China
- Chinese Evidence-Based Medicine Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| |
Collapse
|
40
|
Patel SH, Saito YD, Li Z, Ramaswamy B, Stiff A, Kassem M, Wesolowski R. A solitary brain metastasis as the only site of recurrence of HR positive, HER2 negative breast cancer: a case report and review of the literature. J Med Case Rep 2021; 15:4. [PMID: 33407851 PMCID: PMC7789360 DOI: 10.1186/s13256-020-02615-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 12/01/2020] [Indexed: 11/22/2022] Open
Abstract
Background Breast cancer is one of the most common causes of brain metastases. However, the presence of isolated central nervous system (CNS) metastatic disease early in the course of disease relapse is a rare event in cases of hormone receptor positive, human epidermal growth factor receptor 2 (HER2) negative breast cancer. Case presentation We summarize the clinical course of a pre-menopausal, 39-year old Caucasian female with history of operable, hormone receptor positive, HER2 negative breast cancer who was initially treated with curative-intend therapy but who unfortunately developed solitary metastatic lesion in the left thalamus. A biopsy of the lesion confirmed the presence of hormone receptor positive, HER2 negative metastatic breast cancer. Patient’s CNS metastases continued to progress without any evidence of metastatic disease outside of the central nervous system and she eventually passed away about 5 years after the date of her initial diagnosis and 18 months following the diagnosis with brain metastasis. Conclusion Based on our case, although rare, patients with treated, operable, hormone receptor positive, HER2 negative breast cancer can present with solitary brain metastasis as the only sign of disease recurrence.
Collapse
Affiliation(s)
- Sandipkumar H Patel
- Division of Medical Oncology, The Ohio State University Wexner Medical Center, 1310D Lincoln Tower, 1800 Cannon Dr., Columbus, OH, 43210, USA
| | - Yoshihito David Saito
- The Queen's Medical Center - West Oahu Queen's Cancer Center - West Oahu, Sullivan Care Center, 91-2127 Fort Weaver Road, Ewa Beach, HI, 96706, USA
| | - Zaibo Li
- Department of Pathology, The Ohio State University, 410 W 10th AveN337B Doan Hall, Columbus, OH, 43210-1267, USA
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, The Ohio State University Wexner Medical Center, 1310D Lincoln Tower, 1800 Cannon Dr., Columbus, OH, 43210, USA.,The Ohio State University Comprehensive Cancer Center, The Ohio State University, 410 W 12th Avenue, Columbus, OH, 43210, USA
| | - Andrew Stiff
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, 410 W 12th Avenue, Columbus, OH, 43210, USA
| | - Mahmoud Kassem
- Division of Medical Oncology, The Ohio State University Wexner Medical Center, 1310D Lincoln Tower, 1800 Cannon Dr., Columbus, OH, 43210, USA
| | - Robert Wesolowski
- Division of Medical Oncology, The Ohio State University Wexner Medical Center, 1310D Lincoln Tower, 1800 Cannon Dr., Columbus, OH, 43210, USA. .,The Ohio State University Comprehensive Cancer Center, The Ohio State University, 410 W 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
41
|
Pellerino A, Internò V, Mo F, Franchino F, Soffietti R, Rudà R. Management of Brain and Leptomeningeal Metastases from Breast Cancer. Int J Mol Sci 2020; 21:E8534. [PMID: 33198331 PMCID: PMC7698162 DOI: 10.3390/ijms21228534] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/13/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
The management of breast cancer (BC) has rapidly evolved in the last 20 years. The improvement of systemic therapy allows a remarkable control of extracranial disease. However, brain (BM) and leptomeningeal metastases (LM) are frequent complications of advanced BC and represent a challenging issue for clinicians. Some prognostic scales designed for metastatic BC have been employed to select fit patients for adequate therapy and enrollment in clinical trials. Different systemic drugs, such as targeted therapies with either monoclonal antibodies or small tyrosine kinase molecules, or modified chemotherapeutic agents are under investigation. Major aims are to improve the penetration of active drugs through the blood-brain barrier (BBB) or brain-tumor barrier (BTB), and establish the best sequence and timing of radiotherapy and systemic therapy to avoid neurocognitive impairment. Moreover, pharmacologic prevention is a new concept driven by the efficacy of targeted agents on macrometastases from specific molecular subgroups. This review aims to provide an overview of the clinical and molecular factors involved in the selection of patients for local and/or systemic therapy, as well as the results of clinical trials on advanced BC. Moreover, insight on promising therapeutic options and potential directions of future therapeutic targets against BBB and microenvironment are discussed.
Collapse
Affiliation(s)
- Alessia Pellerino
- Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; (F.M.); (F.F.); (R.S.); (R.R.)
| | - Valeria Internò
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, 70121 Bari, Italy;
| | - Francesca Mo
- Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; (F.M.); (F.F.); (R.S.); (R.R.)
| | - Federica Franchino
- Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; (F.M.); (F.F.); (R.S.); (R.R.)
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; (F.M.); (F.F.); (R.S.); (R.R.)
| | - Roberta Rudà
- Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; (F.M.); (F.F.); (R.S.); (R.R.)
- Department of Neurology, Castelfranco Veneto and Treviso Hospital, 31100 Treviso, Italy
| |
Collapse
|
42
|
Chan WL, Lam TC, Lam KO, Luk MY, Kai-Cheong RN, Kwong LWD. Local and systemic treatment for HER2-positive breast cancer with brain metastases: a comprehensive review. Ther Adv Med Oncol 2020; 12:1758835920953729. [PMID: 32973930 PMCID: PMC7493232 DOI: 10.1177/1758835920953729] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/06/2020] [Indexed: 12/21/2022] Open
Abstract
The management of human epidermal growth factor receptor (HER2)-positive breast cancer has improved over the past decade. However, despite improvements in systemic control, a substantial proportion of patients with advanced HER2-positive breast cancer suffer from central nervous system metastases and even intracranial progression after aggressive local treatment. There is paucity of data and no consensus on the systemic therapies for patients with intracranial progression. This review discusses both local and systemic treatments for HER2-positive breast cancer with brain metastases with a special focus on the response of central nervous system metastases. A recommended practical treatment algorithm to guide physicians in selecting the most appropriate anti-HER2 therapy for their patients is suggested.
Collapse
Affiliation(s)
- Wing-Lok Chan
- Department of Clinical Oncology, Queen Mary Hospital, 1/F Professorial Block, 102 Pokfulam Road, Hong Kong
| | - Tai-Chung Lam
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong SAR
| | - Ka-On Lam
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong SAR
| | - Mai-Yee Luk
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong SAR
| | | | - Lai-Wan Dora Kwong
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong SAR
| |
Collapse
|
43
|
Abstract
Metastasis of cancer cells to the brain occurs frequently in patients with certain subtypes of breast cancer. In particular, patients with HER2-positive or triple-negative breast cancer are at high risk for the development of brain metastases. Despite recent advances in the treatment of primary breast tumors, the prognosis of breast cancer patients with brain metastases remains poor. A better understanding of the molecular and cellular mechanisms underlying brain metastasis might be expected to lead to improvements in the overall survival rate for these patients. Recent studies have revealed complex interactions between metastatic cancer cells and their microenvironment in the brain. Such interactions result in the activation of various signaling pathways related to metastasis in both cancer cells and cells of the microenvironment including astrocytes and microglia. In this review, we focus on such interactions and on their role both in the metastatic process and as potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Mari Hosonaga
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Breast Medical Oncology Department, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yoshimi Arima
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
44
|
Soffietti R, Ahluwalia M, Lin N, Rudà R. Management of brain metastases according to molecular subtypes. Nat Rev Neurol 2020; 16:557-574. [PMID: 32873927 DOI: 10.1038/s41582-020-0391-x] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2020] [Indexed: 12/25/2022]
Abstract
The incidence of brain metastases has markedly increased in the past 20 years owing to progress in the treatment of malignant solid tumours, earlier diagnosis by MRI and an ageing population. Although local therapies remain the mainstay of treatment for many patients with brain metastases, a growing number of systemic options are now available and/or are under active investigation. HER2-targeted therapies (lapatinib, neratinib, tucatinib and trastuzumab emtansine), alone or in combination, yield a number of intracranial responses in patients with HER2-positive breast cancer brain metastases. New inhibitors are being investigated in brain metastases from ER-positive or triple-negative breast cancer. Several generations of EGFR and ALK inhibitors have shown activity on brain metastases from EGFR and ALK mutant non-small-cell lung cancer. Immune-checkpoint inhibitors (ICIs) hold promise in patients with non-small-cell lung cancer without druggable mutations and in patients with triple-negative breast cancer. The survival of patients with brain metastases from melanoma has substantially improved after the advent of BRAF inhibitors and ICIs (ipilimumab, nivolumab and pembrolizumab). The combination of targeted agents or ICIs with stereotactic radiosurgery could further improve the response rates and survival but the risk of radiation necrosis should be monitored. Advanced neuroimaging and liquid biopsy will hopefully improve response evaluation.
Collapse
Affiliation(s)
- Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy.
| | - Manmeet Ahluwalia
- Burkhardt Brain Tumor and Neuro-Oncology Center, Taussig Center Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nancy Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Roberta Rudà
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| |
Collapse
|
45
|
Abstract
Antibodies and antibody fragments have found wide application for therapeutic and diagnostic purposes. Single-domain antibody fragments, also known as 'heavy-chain variable domains' or 'nanobodies', are a recent addition to the toolbox. Discovered some 30 years ago, nanobodies are the smallest antibody-derived fragments that retain antigen-binding properties. Their small size, stability, specificity, affinity and ease of manufacture make them appealing for use as imaging agents in the laboratory and the clinic. With the recent surge in immunotherapeutics and the success of cancer immunotherapy, it is important to be able to image immune responses and cancer biomarkers non-invasively to allocate resources and guide the best possible treatment of patients with cancer. This article reviews recent advances in the application of nanobodies as cancer imaging agents. While much work has been done in preclinical models, first-in-human applications are beginning to show the value of nanobodies as imaging agents.
Collapse
Affiliation(s)
- M. Rashidian
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - H. Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
46
|
Xu B, Hu X, Feng J, Geng C, Jin F, Li H, Li M, Li Q, Liao N, Liu D, Liu J, Liu Q, Lu J, Liu Z, Ma F, Ouyang Q, Pan Y, Shen K, Sun T, Teng Y, Tong Z, Wang B, Wang H, Wang S, Wang S, Wang T, Wang X, Wang X, Wang Y, Wang Z, Wu J, Yan M, Yang J, Yin Y, Yuan P, Zhang J, Zhang P, Zhang Q, Zheng H. Chinese expert consensus on the clinical diagnosis and treatment of advanced breast cancer (2018). Cancer 2020; 126 Suppl 16:3867-3882. [PMID: 32710660 DOI: 10.1002/cncr.32832] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 12/18/2019] [Indexed: 12/19/2022]
|
47
|
Sperduto PW, Mesko S, Li J, Cagney D, Aizer A, Lin NU, Nesbit E, Kruser TJ, Chan J, Braunstein S, Lee J, Kirkpatrick JP, Breen W, Brown PD, Shi D, Shih HA, Soliman H, Sahgal A, Shanley R, Sperduto W, Lou E, Everett A, Boggs DH, Masucci L, Roberge D, Remick J, Plichta K, Buatti JM, Jain S, Gaspar LE, Wu CC, Wang TJC, Bryant J, Chuong M, Yu J, Chiang V, Nakano T, Aoyama H, Mehta MP. Beyond an Updated Graded Prognostic Assessment (Breast GPA): A Prognostic Index and Trends in Treatment and Survival in Breast Cancer Brain Metastases From 1985 to Today. Int J Radiat Oncol Biol Phys 2020; 107:334-343. [PMID: 32084525 PMCID: PMC7276246 DOI: 10.1016/j.ijrobp.2020.01.051] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 02/02/2023]
Abstract
PURPOSE Brain metastases are a common sequelae of breast cancer. Survival varies widely based on diagnosis-specific prognostic factors (PF). We previously published a prognostic index (Graded Prognostic Assessment [GPA]) for patients with breast cancer with brain metastases (BCBM), based on cohort A (1985-2007, n = 642), then updated it, reporting the effect of tumor subtype in cohort B (1993-2010, n = 400). The purpose of this study is to update the Breast GPA with a larger contemporary cohort (C) and compare treatment and survival across the 3 cohorts. METHODS AND MATERIALS A multi-institutional (19), multinational (3), retrospective database of 2473 patients with breast cancer with newly diagnosed brain metastases (BCBM) diagnosed from January 1, 2006, to December 31, 2017, was created and compared with prior cohorts. Associations of PF and treatment with survival were analyzed. Kaplan-Meier survival estimates were compared with log-rank tests. PF were weighted and the Breast GPA was updated such that a GPA of 0 and 4.0 correlate with the worst and best prognoses, respectively. RESULTS Median survival (MS) for cohorts A, B, and C improved over time (from 11, to 14 to 16 months, respectively; P < .01), despite the subtype distribution becoming less favorable. PF significant for survival were tumor subtype, Karnofsky Performance Status, age, number of BCBMs, and extracranial metastases (all P < .01). MS for GPA 0 to 1.0, 1.5-2.0, 2.5-3.0, and 3.5-4.0 was 6, 13, 24, and 36 months, respectively. Between cohorts B and C, the proportion of human epidermal receptor 2 + subtype decreased from 31% to 18% (P < .01) and MS in this subtype increased from 18 to 25 months (P < .01). CONCLUSIONS MS has improved modestly but varies widely by diagnosis-specific PF. New PF are identified and incorporated into an updated Breast GPA (free online calculator available at brainmetgpa.com). The Breast GPA facilitates clinical decision-making and will be useful for stratification of future clinical trials. Furthermore, these data suggest human epidermal receptor 2-targeted therapies improve clinical outcomes in some patients with BCBM.
Collapse
Affiliation(s)
- Paul W Sperduto
- Minneapolis Radiation Oncology & University of Minnesota Gamma Knife Center, Minneapolis, Minnesota.
| | | | - Jing Li
- MD Anderson Cancer Center, Houston, Texas
| | | | - Ayal Aizer
- Dana Farber Cancer Institute, Boston, Massachusetts
| | - Nancy U Lin
- Dana Farber Cancer Institute, Boston, Massachusetts
| | | | | | - Jason Chan
- University of California San Francisco, San Francisco, California
| | - Steve Braunstein
- University of California San Francisco, San Francisco, California
| | | | | | | | | | - Diana Shi
- Massachusetts General Hospital, Massachusetts, Boston, Massachusetts
| | - Helen A Shih
- Massachusetts General Hospital, Massachusetts, Boston, Massachusetts
| | - Hany Soliman
- Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Canada
| | - Arjun Sahgal
- Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Canada
| | | | | | - Emil Lou
- University of Minnesota, Minneapolis, Minnesota
| | | | | | - Laura Masucci
- Centre Hospitalier de l' Université de Montréal, Montreal, Quebec, Canada
| | - David Roberge
- Centre Hospitalier de l' Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | - James Yu
- Yale University, New Haven, Connecticut
| | | | | | | | | |
Collapse
|
48
|
Chen Q, Ouyang D, Anwar M, Xie N, Wang S, Fan P, Qian L, Chen G, Zhou E, Guo L, Gu X, Ding B, Yang X, Liu L, Deng C, Xiao Z, Li J, Wang Y, Zeng S, Hu J, Zhou W, Qiu B, Wang Z, Weng J, Liu M, Li Y, Tang T, Wang J, Zhang H, Dai B, Tang W, Wu T, Xiao M, Li X, Liu H, Li L, Yi W, Ouyang Q. Effectiveness and Safety of Pyrotinib, and Association of Biomarker With Progression-Free Survival in Patients With HER2-Positive Metastatic Breast Cancer: A Real-World, Multicentre Analysis. Front Oncol 2020; 10:811. [PMID: 32528890 PMCID: PMC7263174 DOI: 10.3389/fonc.2020.00811] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/24/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Pyrotinib, an irreversible pan-ERBB inhibitor, has shown promising antitumour activity, and acceptable tolerability. This research was conducted to evaluate the actual use and effectiveness of pyrotinib in China, therefore, contributed to solve the problem of real-world data scarcity. Methods: In this retrospective study, 168 patients who received pyrotinib treatment for HER2-positive metastatic breast cancer (MBC) in Hunan Province from June 2018 to August 2019 were included. Progression-free survival (PFS), tumor mutation burden (TMB), and drug-related adverse events (AEs) after pyrotinib administration were analyzed. Results: The median PFS (mPFS) time in the 168 participants was 8.07 months. The mPFS times in patients with pyrotinib in second-line therapy (n = 65) and third-or-higher-line therapy (n = 94) were 8.10 months and 7.60 months, respectively. Patients with brain metastases achieved 8.80 months mPFS time. In patients with pyrotinib in third-or-higher-line therapy, patients who had previously used lapatinib still got efficacy but showed a shorter mPFS time (6.43 months) than patients who had not (8.37 months). TMB was measured in 28 patients, K-M curve (P = 0.0024) and Multivariate Cox analysis (P = 0.0176) showed a significant negative association between TMB and PFS. Diarrhea occurred in 98.2% of participants (in any grade) and 19.6% in grade 3-4 AEs. Conclusion: Pyrotinib is highly beneficial to second-or-higher-line patients or HER2-positive MBC patients with brain metastases. Pyrotinib seems to be a feasible strategy both in combination of chemotherapeutic drugs or as a replacement of lapatinib if diseases progressed. TMB could be a potential predictor for evaluating pyrotinib's effectiveness in HER2-positive MBC.
Collapse
Affiliation(s)
- Qitong Chen
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Dengjie Ouyang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Munawar Anwar
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ning Xie
- Department of Breast Internal Medicine, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shouman Wang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Peizhi Fan
- Department of Breast and Thyroid Surgery, Hunan Provincial People's Hospital, Changsha, China
| | - Liyuan Qian
- Department of Breast and Thyroid Surgery, Central South University Third Xiangya Hospital, Changsha, China
| | - Gannong Chen
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Enxiang Zhou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lei Guo
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaowen Gu
- Department of Breast and Thyroid Surgery, Hunan Provincial People's Hospital, Changsha, China
| | - Boni Ding
- Department of Breast and Thyroid Surgery, Central South University Third Xiangya Hospital, Changsha, China
| | - Xiaohong Yang
- Department of Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Cental South University, Changsha, China
| | - Liping Liu
- Department of Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Cental South University, Changsha, China
| | - Chao Deng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Xiao
- Department of Breast Internal Medicine, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jing Li
- Department of Breast Medical Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Cental South University, Changsha, China
| | - Yunqi Wang
- Department of Traditional Chinese Medicine, The Affiliated Cancer Hospital of Xiangya School of Medicine, Cental South University, Changsha, China
| | - Shan Zeng
- Department of Internal Medicine - Oncology, Xiangya Hospital Central South University, Changsha, China
| | - Jinhui Hu
- Department of Breast Surgery, The First Hospital Hunan University of Chinese Medicine, Changsha, China
| | - Wei Zhou
- Department of Breast and Thyroid Surgery, The Affiliated ZhuZhou Hospital of Xiangya School of Medicine Central South University, Zhuzhou, China
| | - Bo Qiu
- Department of Oncology, The Affiliated ZhuZhou Hospital of Xiangya School of Medicine Central South University, Zhuzhou, China
| | - Zhongming Wang
- Department of Breast Surgery, The Third People's Hospital of Yongzhou, Yongzhou, China
| | - Jie Weng
- Department of Oncology, The First People's Hospital of Yueyang, Yueyang, China
| | - Mingwen Liu
- Department of Breast and Thyroid Surgery, The First People's Hospital of Xiangtan City, Xiangtan, China
| | - Yi Li
- Department of Oncology, The Third People's Hospital of Changde, Changde, China
| | - Tiegang Tang
- Department of Oncology, Xiangtan Central Hospital, Xiangtan, China
| | - Jianguo Wang
- Department of General Surgery, Xiangtan Central Hospital, Xiangtan, China
| | - Hui Zhang
- Department of Oncology, Central Hospital of Shaoyang, Shaoyang, China
| | - Bin Dai
- Department of Breast and Thyroid Surgery, Central Hospital of Shaoyang, Shaoyang, China
| | - Wuping Tang
- Department of Breast Surgery, Shaoyang Hospital of Traditional Chinese Medicine, Shaoyang, China
| | - Tao Wu
- Department of Oncology, The First People's Hospital of Changde, Changde, China
| | - Maoliang Xiao
- Department of Oncology, The Third Hospital of Hunan University of Chinese Medicine, Zhuzhou, China
| | - Xiantao Li
- Department of Oncology, The Central Hospital of Yiyang, Yiyang, China
| | - Hailong Liu
- Department of Internal Medicine - Oncology, The First People's Hospital of Chenzhou, Chenzhou, China
| | - Lai Li
- Department of Breast and Thyroid Surgery, The People's Hospital of Xiangtan County, Xiangtan, China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Quchang Ouyang
- Department of Breast Internal Medicine, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
49
|
Preclinical Targeted α- and β --Radionuclide Therapy in HER2-Positive Brain Metastasis Using Camelid Single-Domain Antibodies. Cancers (Basel) 2020; 12:cancers12041017. [PMID: 32326199 PMCID: PMC7226418 DOI: 10.3390/cancers12041017] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022] Open
Abstract
HER2-targeted therapies have drastically improved the outcome for breast cancer patients. However, when metastasis to the brain is involved, current strategies fail to hold up to the same promise. Camelid single-domain antibody-fragments (sdAbs) have been demonstrated to possess favorable properties for detecting and treating cancerous lesions in vivo using different radiolabeling methods. Here we evaluate the anti-HER2 sdAb 2Rs15d, coupled to diagnostic γ- and therapeutic α- and β−-emitting radionuclides for the detection and treatment of HER2pos brain lesions in a preclinical setting. 2Rs15d was radiolabeled with 111In, 225Ac and 131I using DTPA- and DOTA-based bifunctional chelators and Sn-precursor of SGMIB respectively and evaluated in orthotopic tumor-bearing athymic nude mice. Therapeutic efficacy as well as systemic toxicity were determined for 131I- and 225Ac-labeled sdAbs and compared to anti-HER2 monoclonal antibody (mAb) trastuzumab in two different HER2pos tumor models. Radiolabeled 2Rs15d showed high and specific tumor uptake in both HER2pos SK-OV-3-Luc-IP1 and HER2pos MDA-MB-231Br brain lesions, whereas radiolabeled trastuzumab was unable to accumulate in intracranial SK-OV-3-Luc-IP1 tumors. Administration of [131I]-2Rs15d and [225Ac]-2Rs15d alone and in combination with trastuzumab showed a significant increase in median survival in 2 tumor models that remained largely unresponsive to trastuzumab treatment alone. Histopathological analysis revealed no significant early toxicity. Radiolabeled sdAbs prove to be promising vehicles for molecular imaging and targeted radionuclide therapy of metastatic lesions in the brain. These data demonstrate the potential of radiolabeled sdAbs as a valuable add-on treatment option for patients with difficult-to-treat HER2pos metastatic cancer.
Collapse
|
50
|
Thulin A, Rönnerman E, Zhang C, De Lara S, Chamalidou C, Schoenfeldt A, Andersson C, Kovács A, Enlund F, Linderholm B. Clinical outcome of patients with brain metastases from breast cancer - A population based study over 21 years. Breast 2020; 50:113-124. [PMID: 32145571 PMCID: PMC7375610 DOI: 10.1016/j.breast.2020.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 01/14/2020] [Accepted: 02/10/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Brain metastases (BM) are a feared progression of breast cancer (BC) with impact on quality of life and survival. Despite improved treatments, it is believed patients suffering from BM are increasing. AIMS To study potential changes in the number of BM, the possible links between BC subgroup and extent of BM with prognosis. To investigate the interval between primary BC/extra cranial recurrence, and diagnosis of BM in the years 1994-2014. PATIENTS AND METHODS Clinical data from 191 patients with BM diagnosed 1994-2014, was retrieved from charts. Primary tumours where re-evaluated histologically. RESULTS There was an increase of BM in 5 years cohorts (1994-99 (n = 9); 2000-04 (n = 36); 2005-09 (n = 60); 2010-14 (n = 86)). We found no difference in the time interval from primary BC to BM but an insignificant increase in time from extra cranial relapse to development of BM in the time periods 1994-2004 and 2005-2014 of 15.5 and 25.0 months (p = 0.0612). Survival after BM was 7 months (95% CI 6-10) with a statistically significant difference between HER2 positive and TNBC with an inferior outcome for the latter (p = 0.018) whilst no differences were present when Luminal BC were compared with HER2 positive BC (p = 0.073). CONCLUSIONS We show an increase of BM over time whilst the time span from primary BC to BM is unchanged supports earlier findings that adjuvant treatments have little preventive function. Time from extra cranial recurrence to BM was prolonged with one year. Patients with TNBC or more advance extent of BM had the shortest survival with BM.
Collapse
Affiliation(s)
- Anna Thulin
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Elisabeth Rönnerman
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Shahin De Lara
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Chaido Chamalidou
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Department of Oncology, SKAS, Sweden
| | - Arnd Schoenfeldt
- Department of Pathology, Norra Älvsborgs Hospital, Trollhättan, Sweden
| | - Carola Andersson
- Diagnostic Center, Kalmar, Regional Hospital of Kalmar County, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Fredrik Enlund
- Diagnostic Center, Kalmar, Regional Hospital of Kalmar County, Sweden
| | - Barbro Linderholm
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|