1
|
Zhang M, Zhou K, Wang Z, Liu T, Stevens LE, Lynce F, Chen WY, Peng S, Xie Y, Zhai D, Chen Q, Shi Y, Shi H, Yuan Z, Li X, Xu J, Cai Z, Guo J, Shao N, Lin Y. A Subpopulation of Luminal Progenitors Secretes Pleiotrophin to Promote Angiogenesis and Metastasis in Inflammatory Breast Cancer. Cancer Res 2024; 84:1781-1798. [PMID: 38507720 PMCID: PMC11148543 DOI: 10.1158/0008-5472.can-23-2640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/19/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024]
Abstract
Inflammatory breast cancer (IBC) is a highly aggressive subtype of breast cancer characterized by rapidly arising diffuse erythema and edema. Genomic studies have not identified consistent alterations and mechanisms that differentiate IBC from non-IBC tumors, suggesting that the microenvironment could be a potential driver of IBC phenotypes. Here, using single-cell RNA sequencing, multiplex staining, and serum analysis in patients with IBC, we identified enrichment of a subgroup of luminal progenitor (LP) cells containing high expression of the neurotropic cytokine pleiotrophin (PTN) in IBC tumors. PTN secreted by the LP cells promoted angiogenesis by directly interacting with the NRP1 receptor on endothelial tip cells located in both IBC tumors and the affected skin. NRP1 activation in tip cells led to recruitment of immature perivascular cells in the affected skin of IBC, which are correlated with increased angiogenesis and IBC metastasis. Together, these findings reveal a role for cross-talk between LPs, endothelial tip cells, and immature perivascular cells via PTN-NRP1 axis in the pathogenesis of IBC, which could lead to improved strategies for treating IBC. SIGNIFICANCE Nonmalignant luminal progenitor cells expressing pleiotrophin promote angiogenesis by activating NRP1 and induce a prometastatic tumor microenvironment in inflammatory breast cancer, providing potential therapeutic targets for this aggressive breast cancer subtype.
Collapse
Affiliation(s)
- Mengmeng Zhang
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kaiwen Zhou
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zilin Wang
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ting Liu
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Laura E Stevens
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Filipa Lynce
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Wendy Y Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sui Peng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yubin Xie
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Duanyang Zhai
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qianjun Chen
- Department of Breast Oncology, Traditional Chinese Medicine Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Yawei Shi
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huijuan Shi
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhongyu Yuan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoping Li
- Department of Breast Oncology, Jiangmen Central Hospital, Jiangmen, China
| | - Juan Xu
- Department of Breast Oncology, Maternal and Child Health Care Hospital of Guangdong Province, Guangzhou, China
| | - Zhenhai Cai
- Department of Breast Oncology, Jieyang People's Hospital, Jieyang, China
| | - Jianping Guo
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Nan Shao
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Lin
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Iwase T, Wang X, Thi Hanh Phi L, Sridhar N, Ueno NT, Lee J. Advances in targets in inflammatory breast cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 384:125-152. [PMID: 38637096 DOI: 10.1016/bs.ircmb.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Affiliation(s)
- Toshiaki Iwase
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; University of Hawaii Cancer Center, Honolulu, HI, United States.
| | - Xiaoping Wang
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lan Thi Hanh Phi
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nithya Sridhar
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; University of Hawaii Cancer Center, Honolulu, HI, United States
| | - Jangsoon Lee
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
3
|
Deng J, Hua L, Bian L, Chen H, Chen L, Cheng H, Dou C, Geng D, Hong T, Ji H, Jiang Y, Lan Q, Li G, Liu Z, Qi S, Qu Y, Shi S, Sun X, Wang H, You Y, Yu H, Yue S, Zhang J, Zhang X, Wang S, Mao Y, Zhong P, Gong Y. Molecular diagnosis and treatment of meningiomas: an expert consensus (2022). Chin Med J (Engl) 2022; 135:1894-1912. [PMID: 36179152 PMCID: PMC9746788 DOI: 10.1097/cm9.0000000000002391] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Indexed: 11/27/2022] Open
Abstract
ABSTRACT Meningiomas are the most common primary intracranial neoplasm with diverse pathological types and complicated clinical manifestations. The fifth edition of the WHO Classification of Tumors of the Central Nervous System (WHO CNS5), published in 2021, introduces major changes that advance the role of molecular diagnostics in meningiomas. To follow the revision of WHO CNS5, this expert consensus statement was formed jointly by the Group of Neuro-Oncology, Society of Neurosurgery, Chinese Medical Association together with neuropathologists and evidence-based experts. The consensus provides reference points to integrate key biomarkers into stratification and clinical decision making for meningioma patients. REGISTRATION Practice guideline REgistration for transPAREncy (PREPARE), IPGRP-2022CN234.
Collapse
Affiliation(s)
- Jiaojiao Deng
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lingyang Hua
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai 200040, China
- Neurosurgical Institute of Fudan University, Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
| | - Liuguan Bian
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hong Chen
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Hongwei Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Changwu Dou
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 750306, China
| | - Dangmurenjiapu Geng
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Hongming Ji
- Department of Neurosurgery, Shanxi Medical University Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Yugang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qing Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Soochow, Jiangsu 215004, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250063, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi 710038, China
| | - Songsheng Shi
- Department of Neurosurgery, Fujian Medical University Affiliated Union Hospital, Fuzhou, Fujian 350001, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Haijun Wang
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Hualin Yu
- Department of Neurosurgery, Kunming Medical University First Affiliated Hospital, Kunming, Yunnan 650032, China
| | - Shuyuan Yue
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jianming Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Xiaohua Zhang
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ping Zhong
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai 200040, China
- Neurosurgical Institute of Fudan University, Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
| | - Ye Gong
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
4
|
Bendavid J, Modesto A. Radiation therapy and antiangiogenic therapy: Opportunities and challenges. Cancer Radiother 2022; 26:962-967. [PMID: 35989153 DOI: 10.1016/j.canrad.2022.06.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/21/2022] [Indexed: 10/15/2022]
Abstract
The importance of tumoral vascularization as a therapeutic target was first described in 1971 by Folkman. Anarchic vascularization in response to tumour hypoxia, especially mediated by vascular endothelial growth factor, represents a major target in the management of many cancers. The contribution of systemic anti-angiogenic treatments including humanized anti-VEGF monoclonal antibodies (bevacizumab) and tyrosine kinase inhibitors, whose effect on vascular normalization and correction of tumour hypoxia has been shown in preclinical studies to be enhancing the effect of radiotherapy. Early trials combining radiotherapy and antiangiogenics with a small number of patients have contradictory results and tend to put into perspective the opportunity that this synergistic association represents. The efficiency found must be tempered by some toxicity described, especially in association with high doses per fraction. The aim of this article is to present the main studies reporting the efficiency and safety of the combination of antiangiogenic drugs and radiotherapy, as well as the expected opportunities.
Collapse
Affiliation(s)
- J Bendavid
- Département de radiothérapie, Gustave-Roussy, 114, rue Édouard-Vaillant, 94805 Villejuif, France.
| | - A Modesto
- Département de radiothérapie, IUCT Oncopole, 1, avenue Irène-Jolio-Curie, 31100 Toulouse, France
| |
Collapse
|
5
|
Radiotherapy in the Management of Non-Metastatic Inflammatory Breast Cancers: A Retrospective Observational Study. Cancers (Basel) 2021; 14:cancers14010107. [PMID: 35008271 PMCID: PMC8750160 DOI: 10.3390/cancers14010107] [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: 11/27/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 11/17/2022] Open
Abstract
(1) Background: Inflammatory breast cancers (IBC) are characterized by a poor prognosis. This retrospective study aims to describe the clinical outcomes of non-metastatic IBC patients treated with a multidisciplinary approach with neo-adjuvant chemotherapy, surgery, and radiotherapy. (2) Methods: This single-center retrospective study included all women patients diagnosed with non-metastatic IBC between January 2010 and January 2018 at the Institut Curie (Paris, France) and treated with neoadjuvant chemotherapy, surgery, and radiotherapy. Overall survival (OS), disease-free survival (DFS), and locoregional free survival (LRRFS) were calculated from the time of diagnosis. Prognostic factors for patient survival were analyzed based on univariate and multivariate regressions. (3) Results: We identified 113 patients with a median age of 51 years. 79.7% had node-positive tumors; triple-negative breast cancers (TNBC) represented 34.6% of the cases. A large majority of patients (91.2%) received adjuvant post-mastectomy while ten patients (8.8%) received preoperative radiotherapy. Non-pathological complete response (non-pCR) was observed in 67.3% of patients. Radiotherapy delivered a median dose of 50 Gy to the breast or the chest wall in 25 fractions. With a median follow-up of 54 months, 5-year OS, DFS and LRRFS were 78% (CI: 70.1-86.8%), 68.1% (59.6-77.7%), and 85.2% (78.4-92.7%), respectively. In multivariate analysis, non-pCR was an adverse prognosis factor for OS, DFS, and LRRFS; pre-operative radiotherapy was an adverse prognosis factor for OS and DFS. Radiation-related adverse events were limited to acute skin toxicity (22% of Grade 2 and 2% of grade 3 dermatitis); no late radiation-induced toxicity was reported. (4) Conclusions: High locoregional control could be achieved with multidisciplinary management of non-metastatic IBC, suggesting the anti-tumor efficacy of radiotherapy in this rare but pejorative clinicopathological presentation. While comparing favorably with historical cohorts, OS and DFS could be potentially improved in the future with the use of new systemic treatments, such as PARP-inhibitors or immunotherapy.
Collapse
|
6
|
Progress for Immunotherapy in Inflammatory Breast Cancer and Emerging Barriers to Therapeutic Efficacy. Cancers (Basel) 2021; 13:cancers13112543. [PMID: 34067257 PMCID: PMC8196819 DOI: 10.3390/cancers13112543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Despite recent advances in the treatment of other breast cancer subtypes, inflammatory breast cancer (IBC) remains a significant clinical challenge, with an overall 5-year survival rate of 39%. Though immunotherapy has shown remarkable efficacy in other difficult-to-treat cancers, such approaches have yet to show substantial therapeutic efficacy in IBC. Here, we summarize the known immune composition of IBC tumors, as well as past and present efforts to advance immunotherapy in the treatment of IBC. Abstract Inflammatory breast cancer (IBC) is a rare and aggressive subtype of breast cancer that carries a particularly poor prognosis. Despite the efficacy of immunotherapy in other difficult to treat forms of breast cancer, progress for immunotherapy in IBC has been difficult. Though immunotherapy has been under clinical investigation in IBC since the 1970s, few approaches have shown significant therapeutic efficacy, and no immunotherapy regimens are currently used in the treatment of IBC. Here, we provide a comprehensive summary of what is known about the immune composition of IBC tumors, clinical and basic science evidence describing the role for immune checkpoints such as PD-L1 in IBC pathobiology, as well as past and present attempts to advance ICIs in the treatment of IBC.
Collapse
|
7
|
Chainitikun S, Espinosa Fernandez JR, Long JP, Iwase T, Kida K, Wang X, Saleem S, Lim B, Valero V, Ueno NT. Pathological complete response of adding targeted therapy to neoadjuvant chemotherapy for inflammatory breast cancer: A systematic review. PLoS One 2021; 16:e0250057. [PMID: 33861773 PMCID: PMC8051801 DOI: 10.1371/journal.pone.0250057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/31/2021] [Indexed: 01/06/2023] Open
Abstract
Background The current use of targeted therapy plus neoadjuvant chemotherapy for inflammatory breast cancer (IBC) is based on data extrapolated from studies in non-IBC. We conducted a systematic review to determine whether neoadjuvant chemotherapy plus targeted therapy results in a higher pathologic complete response (pCR) rate than neoadjuvant chemotherapy alone in patients with IBC. Method and findings This systematic review was registered in the PROSPERO register with registration number CRD42018089465. We searched MEDLINE & PubMed, EMBASE, and EBSCO from December 1998 through July 2020. All English-language clinical studies, both randomized and non-randomized, that evaluated neoadjuvant systemic treatment with or without targeted therapy before definitive surgery and reported the pCR results of IBC patients. First reviewer extracted data and assessed the risk of bias using the Risk of Bias In Non-randomized Studies of Interventions tool. Second reviewer confirmed the accuracy. Studies were divided into 3 groups according to systemic treatment: chemotherapy with targeted therapy, chemotherapy alone, and high-dose chemotherapy with hematopoietic stem cell support (HSCS). Of 995 screened studies, 23 with 1,269 IBC patients met the inclusion criteria. For each of the 3 groups of studies, we computed a weighted average of the pCR rates across all studies with confidence interval (CI). The weighted averages (95% CIs) were as follows: chemotherapy with targeted therapy, 31.6% (26.4%-37.3%), chemotherapy alone, 13.0% (10.3%-16.2%), and high-dose chemotherapy with HSCS, 23.0% (18.7%-27.7%). The high pCR by targeted therapy group came from anti-HER2 therapy, 54.4% (44.3%-64.0%). Key limitations of this study included no randomized clinical studies that included only IBC patients. Conclusion Neoadjuvant chemotherapy plus targeted therapy is more effective than neoadjuvant chemotherapy alone for IBC patients. These findings support current IBC standard practice in particular the use of anti-HER2 targeted therapy.
Collapse
Affiliation(s)
- Sudpreeda Chainitikun
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jose Rodrigo Espinosa Fernandez
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - James P. Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Toshiaki Iwase
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Kumiko Kida
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Xiaoping Wang
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sadia Saleem
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Bora Lim
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Vicente Valero
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Naoto T. Ueno
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
8
|
Chainitikun S, Saleem S, Lim B, Valero V, Ueno NT. Update on systemic treatment for newly diagnosed inflammatory breast cancer. J Adv Res 2021; 29:1-12. [PMID: 33842000 PMCID: PMC8020152 DOI: 10.1016/j.jare.2020.08.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/31/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
Background Inflammatory breast cancer (IBC) is a rare and aggressive disease, accounting for 2-4% of new cases of breast cancer. Owing to its aggressive nature, IBC represent approximately 8-10% of breast cancer deaths. Management of IBC requires a multidisciplinary team for decision-making involving a composite of systemic treatment, surgery, and radiation, or "Trimodality Treatment." Because of the rarity of the disease, systemic therapy of IBC traditionally has been extrapolated from non-IBC clinical trials. Aim of Review The purpose of this review is to provide an overview of the development of systemic treatment of IBC from the past to the present by focusing on IBC clinical trials, including chemotherapy and targeted therapies. Key Scientific Concepts of Review We discuss their effects on pathologic complete response (pCR) and survival outcomes, the predictive markers, and the adverse events of these therapies. Further, we summarized the current standard treatment stratified by molecular subtypes based on clinical data. Finally, we discuss the future trend of systemic therapy, including immunotherapy and ongoing IBC clinical trials.
Collapse
Affiliation(s)
- Sudpreeda Chainitikun
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Sadia Saleem
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Bora Lim
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Vicente Valero
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Naoto T. Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
9
|
Overview of New Treatments with Immunotherapy for Breast Cancer and a Proposal of a Combination Therapy. Molecules 2020; 25:molecules25235686. [PMID: 33276556 DOI: 10.3390/molecules25235686] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 01/08/2023] Open
Abstract
According to data from the U.S. National Cancer Institute, cancer is one of the leading causes of death worldwide with approximately 14 million new cases and 8.2 million cancer-related deaths in 2018. More than 60% of the new annual cases in the world occur in Africa, Asia, Central America, and South America, with 70% of cancer deaths in these regions. Breast cancer is the most common cancer in women, with 266,120 new cases in American women and an estimated 40,920 deaths for 2018. Approximately one in six women diagnosed with breast cancer will die in the coming years. Recently, novel therapeutic strategies have been implemented in the fight against breast cancer, including molecules able to block signaling pathways, an inhibitor of poly [ADP-ribose] polymerase (PARP), growth receptor blocker antibodies, or those that reactivate the immune system by inhibiting the activities of inhibitory receptors like cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death protein 1 (PD-1). However, novel targets include reactivating the Th1 immune response, changing tumor microenvironment, and co-activation of other components of the immune response such as natural killer cells and CD8+ T cells among others. In this article, we review advances in the treatment of breast cancer focused essentially on immunomodulatory drugs in targeted cancer therapy. Based on this knowledge, we formulate a proposal for the implementation of combined therapy using an extracorporeal immune response reactivation model and cytokines plus modulating antibodies for co-activation of the Th1- and natural killer cell (NK)-dependent immune response, either in situ or through autologous cell therapy. The implementation of "combination immunotherapy" is new hope in breast cancer treatment. Therefore, we consider the coordinated activation of each cell of the immune response that would probably produce better outcomes. Although more research is required, the results recently achieved by combination therapy suggest that for most, if not all, cancer patients, this tailored therapy may become a realistic approach in the near future.
Collapse
|
10
|
Alba-Bernal A, Lavado-Valenzuela R, Domínguez-Recio ME, Jiménez-Rodriguez B, Queipo-Ortuño MI, Alba E, Comino-Méndez I. Challenges and achievements of liquid biopsy technologies employed in early breast cancer. EBioMedicine 2020; 62:103100. [PMID: 33161226 PMCID: PMC7670097 DOI: 10.1016/j.ebiom.2020.103100] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/01/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is the most common cancer type in women worldwide and its early detection is crucial to curing the disease. Tissue biopsy, currently the method of choice to obtain tumour molecular information, is invasive and might be affected by tumour heterogeneity rendering it incapable to portray the complete molecular picture. Liquid biopsy permits to study disease features in a more comprehensive manner by sampling biofluids and extracting tumour components such as circulating-tumour DNA (ctDNA), circulating-tumour cells (CTCs), and/or circulating-tumour RNA (ctRNA) amongst others in a monitoring-compatible manner. In this review, we describe the recent progress in the utilization of the circulating tumour components using early breast cancer samples. We review the most important analytes and technologies employed for their study.
Collapse
Affiliation(s)
- Alfonso Alba-Bernal
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria. The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), Málaga 29010, Spain
| | - Rocío Lavado-Valenzuela
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria. The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), Málaga 29010, Spain
| | - María Emilia Domínguez-Recio
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria. The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), Málaga 29010, Spain
| | - Begoña Jiménez-Rodriguez
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria. The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), Málaga 29010, Spain
| | - María Isabel Queipo-Ortuño
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria. The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), Málaga 29010, Spain
| | - Emilio Alba
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria. The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), Málaga 29010, Spain.
| | - Iñaki Comino-Méndez
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria. The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), Málaga 29010, Spain.
| |
Collapse
|
11
|
Bertucci A, Bertucci F, Zemmour C, Lerebours F, Pierga JY, Levy C, Dalenc F, Grenier J, Petit T, Berline M, Gonçalves A. PELICAN-IPC 2015-016/Oncodistinct-003: A Prospective, Multicenter, Open-Label, Randomized, Non-Comparative, Phase II Study of Pembrolizumab in Combination With Neo Adjuvant EC-Paclitaxel Regimen in HER2-Negative Inflammatory Breast Cancer. Front Oncol 2020; 10:575978. [PMID: 33330051 PMCID: PMC7732675 DOI: 10.3389/fonc.2020.575978] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/28/2020] [Indexed: 12/31/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a highly aggressive entity with a poor outcome and relative resistance to treatment. Despite progresses achieved during the last decades, the survival remains significantly lower than non-IBC. Recent clinical trials assessing PD-1/PD-L1 inhibitors showed promising results in non-IBC. Pembrolizumab, an anti-PD-1 monoclonal antibody, revolutionized the treatment of different cancers. Several recent studies suggested a potential interest of targeting the immune system in IBC by revealing a more frequent PD-L1 expression and an enriched immune microenvironment when compared with non-IBC. Here, we describe the rationale and design of PELICAN-IPC 2015-016/Oncodistinct-003 trial, an open-label, randomized, non-comparative, phase II study assessing efficacy, and safety of pembrolizumab in combination with anthracycline-containing neoadjuvant chemotherapy in HER2-negative IBC. The trial is ongoing. The primary endpoint is the pCR rate (ypT0/Tis, ypN0) in overall population and the co-primary endpoint is safety profile during a run-in phase. Key secondary objectives include tolerability, invasive disease-free, event-free and overall survivals, as well as collection of tumor and blood samples for translational research. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ (NCT03515798).
Collapse
Affiliation(s)
- Alexandre Bertucci
- Département d’Oncologie Médicale, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
| | - François Bertucci
- Département d’Oncologie Médicale, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
- Aix-Marseille Université, Faculté de Médecine, Marseille, France
| | - Christophe Zemmour
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
| | | | - Jean-Yves Pierga
- Département d’Oncologie Médicale, Institut Curie, Saint-Cloud, France
- Département d’Oncologie Médicale, Institut Curie, Université de Paris, Paris, France
| | - Christelle Levy
- Breast Cancer Unit, François Baclesse Cancer Centre, Caen, France
| | - Florence Dalenc
- Département d’Oncologie Médicale, Institut Claudius Regaud, Institut Universitaire du Cancer, Oncopole Toulouse, France
| | - Julien Grenier
- Département d’Oncologie Médicale, Institut Sainte Catherine, Avignon, France
| | - Thierry Petit
- Département d’Oncologie Médicale, Centre Paul-Strauss, Strasbourg, France
| | - Marguerite Berline
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
| | - Anthony Gonçalves
- Département d’Oncologie Médicale, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Marseille, France
- Aix-Marseille Université, Faculté de Médecine, Marseille, France
- Oncodistinct Network, Bruxelles, Belgium
| |
Collapse
|
12
|
The Role of Circulating Tumor Cells in Breast Cancer and Implications for Radiation Treatment Decisions. Int J Radiat Oncol Biol Phys 2020; 109:44-59. [PMID: 32882354 DOI: 10.1016/j.ijrobp.2020.08.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/05/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Tumor biomarkers are used routinely in oncology to assign risk categorization, screen and assist in diagnosis of malignancy, allow for prognostication and prediction of outcomes and treatment response, and allow for monitoring of patients after treatment completion. Although tissue-based biomarkers have a long history of use, the emergence of liquid-based biomarkers, including circulating tumor cells (CTCs), may soon revolutionize the management of patients with cancer. Here, we review the discovery of CTCs and their role as prognostic and predictive biomarkers, with an emphasis on breast cancer. We discuss the platforms for CTC enumeration and focus on studies using the only US Food and Drug Administration-approved platform for CTC enumeration (CellSearch). In addition, we examine the role of CTCs in women with metastatic, inflammatory, and nonmetastatic breast cancer, as well as the clinical evidence for their use as a surrogate for radiation treatment response as well as surveillance after treatment. Finally, we conclude by investigating ongoing clinical studies assessing CTCs as radiation response predictors and discuss unanswered questions.
Collapse
|
13
|
Targeting Signaling Pathways in Inflammatory Breast Cancer. Cancers (Basel) 2020; 12:cancers12092479. [PMID: 32883032 PMCID: PMC7563157 DOI: 10.3390/cancers12092479] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 12/17/2022] Open
Abstract
Inflammatory breast cancer (IBC), although rare, is the most aggressive type of breast cancer. Only 2-4% of breast cancer cases are classified as IBC, but-owing to its high rate of metastasis and poor prognosis-8% to 10% of breast cancer-related mortality occur in patients with IBC. Currently, IBC-specific targeted therapies are not available, and there is a critical need for novel therapies derived via understanding novel targets. In this review, we summarize the biological functions of critical signaling pathways in the progression of IBC and the preclinical and clinical studies of targeting these pathways in IBC. We also discuss studies of crosstalk between several signaling pathways and the IBC tumor microenvironment.
Collapse
|
14
|
Lerebours F, Vacher S, Guinebretiere JM, Rondeau S, Caly M, Gentien D, Van Laere S, Bertucci F, de la Grange P, Bièche L, Liang X, Callens C. Hemoglobin overexpression and splice signature as new features of inflammatory breast cancer? J Adv Res 2020; 28:77-85. [PMID: 33364047 PMCID: PMC7753232 DOI: 10.1016/j.jare.2020.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/30/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction Inflammatory Breast Cancer (IBC) is the most aggressive form of breast carcinoma characterized by rapid onset of inflammatory signs and its molecular fingerprint has not yet been elucidated. Objectives The objective of this study was to detect both gene expression levels and alternate RNA splice variants specific for IBC. Methods W e performed splice-sensitive array profiling using Affymetrix Exon Array and quantitative RT-PCR analyses in 177 IBC compared to 183 non-IBC. We also assessed the prognostic value of the identified candidate genes and splice variants. Results A 5-splice signature (HSPA8, RPL10, RPL4, DIDO1 and EVL) was able to distinguish IBC from non-IBC tumors (p<10-7). This splice signature was associated with poor metastasis-free survival in hormone receptor-negative non-IBC (p=0.02), but had no prognostic value in IBC. PAM analysis of dysregulated genes in IBC compared to non-IBC identified a 10-gene signature highly predictive of IBC phenotype and conferring a poor prognosis in non-IBC. The genes most commonly upregulated in IBC were 3 hemoglobin genes able to reliably discriminate IBC from non-IBC (p<10-4). Hb protein expression in epithelial breast tumor cells was confirmed by immunohistochemistry. Conclusion IBC has a specific spliced transcript profile and is characterized by hemoglobin gene overexpression that should be investigated in further functional studies.
Collapse
Affiliation(s)
- F Lerebours
- Département d'Oncologie Médicale, Institut Curie-Hôpital René Huguenin, Saint-Cloud, France
| | - S Vacher
- Service de Génétique, Unité de Pharmacogénomique, Institut Curie, Université Paris Sciences et Lettres, Paris, France
| | - J M Guinebretiere
- Département de Biopathologie, Institut Curie-Hôpital René Huguenin, Saint-Cloud, France
| | - S Rondeau
- Service de Génétique, Unité de Pharmacogénomique, Institut Curie, Université Paris Sciences et Lettres, Paris, France
| | - M Caly
- Département de Biopathologie, Institut Curie-Hôpital René Huguenin, Saint-Cloud, France
| | - D Gentien
- Plateforme de Génomique, Département de Recherche Translationnelle, Institut Curie, Université Paris Sciences et Lettres, Paris, France
| | - S Van Laere
- Translational Cancer Research Unit Antwerp, General Hospital Sint Augustinus, Wilrijk, Belgium
| | - F Bertucci
- Département d'Oncologie Médicale, Institut Paoli-Calmettes, Marseille, France
| | | | - L Bièche
- Service de Génétique, Unité de Pharmacogénomique, Institut Curie, Université Paris Sciences et Lettres, Paris, France.,INSERM U1016, Paris Descartes University, Faculty of Pharmaceutical and Biological Sciences, Paris, France
| | - X Liang
- Service de Génétique, Unité de Pharmacogénomique, Institut Curie, Université Paris Sciences et Lettres, Paris, France.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - C Callens
- Service de Génétique, Unité de Pharmacogénomique, Institut Curie, Université Paris Sciences et Lettres, Paris, France
| |
Collapse
|
15
|
Martos T, Casadevall D, Albanell J. Circulating Tumor Cells: Applications for Early Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1220:135-146. [PMID: 32304084 DOI: 10.1007/978-3-030-35805-1_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Breast cancer is the most common malignancy among women. Most of breast cancer patients are diagnosed in early stages and will be treated with curative intent. Despite this, some patients will relapse. The identification of patients at high risk remains an important challenge. CTCs can be useful to identify this patients, to assess tumor dynamics and to monitoring therapy. There is definitive evidence on the prognostic role of CTCs in early breast cancer (eBC) but its clinical utility in daily practice is still lacking. We have to take into consideration that the studies published to date mainly evaluated the presence of CTC based on the expression of epithelial surface markers. Future studies need to overcome this limitation and important advances in technical methods can assess CTCs and capture the heterogeneity of the tumor landscape. It is also tempting to speculate that CTCs may also provide complementary information on the interplay of tumor cells with the immune system. The combination of different methods to detect tumoral disease by liquid biopsy may provide new ways to personalize in an unprecedented manner the management of patients with eBC.
Collapse
Affiliation(s)
- Tamara Martos
- Servei d'Oncologia Mèdica, Hospital del Mar, Barcelona, Spain
| | - David Casadevall
- Servei d'Oncologia Mèdica, Hospital del Mar, Barcelona, Spain.,Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Joan Albanell
- Servei d'Oncologia Mèdica, Hospital del Mar, Barcelona, Spain. .,Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain. .,CEXS Department, Pompeu Fabra University, Barcelona, Spain. .,CIOCC HM Delfos, Barcelona, Spain.
| |
Collapse
|
16
|
Vasseur A, Cabel L, Tredan O, Chevrier M, Dubot C, Lorgis V, Jacot W, Goncalves A, Debled M, Levy C, Ferrero JM, Jouannaud C, Luporsi E, Mouret-Reynier MA, Dalenc F, Lemonnier J, Savignoni A, Tanguy ML, Bidard FC, Pierga JY. Prognostic value of CEC count in HER2-negative metastatic breast cancer patients treated with bevacizumab and chemotherapy: a prospective validation study (UCBG COMET). Angiogenesis 2019; 23:193-202. [PMID: 31773439 DOI: 10.1007/s10456-019-09697-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/16/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Proof of concept studies has reported that circulating endothelial cell (CEC) count may be associated with the outcome of HER2-negative metastatic breast cancer (mBC) patients treated by chemotherapy and the anti-VEGF antibody bevacizumab. We report the results obtained in an independent prospective validation cohort (COMET study, NCT01745757). METHODS The main baseline criteria were HER2-negative mBC, performance status 0-2 and no prior chemotherapy for metastatic disease. CECs were detected by CellSearch® from 4 ml of blood at baseline and after 4 weeks of weekly paclitaxel and bevacizumab therapy. CEC counts (considered both as a continuous variable and using the previously described 20 CEC/4 ml cutoff) were associated with clinical characteristics and progression-free survival (PFS). RESULTS CEC count was obtained in 251 patients at baseline and in 207 patients at 4 weeks. Median baseline CEC count was 22 CEC/4 ml (range 0-2231). Baseline CEC counts were associated with performance status (p = 0.02). No statistically significant change in CEC counts was observed between baseline and 4 weeks of therapy. High baseline CEC count was associated with shorter PFS in univariate and multivariate analyses (continuous: p < 0.001; dichotomized: HR 1.52, 95% CI [1.15-2.02], p = 0.004). CEC counts at 4 weeks had no prognostic impact. CONCLUSION This study confirms that CEC count may be associated with the outcome of mBC patients treated with chemotherapy and bevacizumab. However, discrepancies with previous reports in terms of both the timing of CEC count and the direction of the prognostic impact warrant further clinical investigation.
Collapse
Affiliation(s)
- Antoine Vasseur
- Department of Medical Oncology, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris & Saint Cloud, France
| | - Luc Cabel
- Department of Medical Oncology, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris & Saint Cloud, France.,UVSQ, Paris-Saclay University, Saint Cloud, France
| | - Olivier Tredan
- Department of Medical Oncology, Leon Berard Center, Lyon, France
| | - Marion Chevrier
- Department of Biostatistics, Institut Curie, PSL Research University, Paris & Saint Cloud, France
| | - Coraline Dubot
- Department of Medical Oncology, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris & Saint Cloud, France
| | - Véronique Lorgis
- Department of Medical Oncology, Georges-François Leclerc Center, Dijon, France
| | - William Jacot
- Department of Medical Oncology, Institut du Cancer de Montpellier (ICM) Val d'Aurelle, Montpellier University, IRCM INSERM U1194, Montpellier, France
| | - Anthony Goncalves
- Aix-Marseille Univ, Inserm U1068, CNRS UMR7258, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Marc Debled
- Department of Medical Oncology, Institut Bergonie, Bordeaux, France
| | - Christelle Levy
- Department of Medical Oncology, François Baclesse Center, Caen, France
| | - Jean-Marc Ferrero
- Department of Medical Oncology, Antoine Lacassagne Center, Nice, France
| | | | - Elisabeth Luporsi
- Department of Medical Oncology, ICL Alexis Vautrin, Vandoeuvre Les Nancy, France
| | | | - Florence Dalenc
- Department of Medical Oncology, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France
| | | | - Alexia Savignoni
- Department of Biostatistics, Institut Curie, PSL Research University, Paris & Saint Cloud, France
| | - Marie-Laure Tanguy
- Department of Biostatistics, Institut Curie, PSL Research University, Paris & Saint Cloud, France
| | - Francois-Clement Bidard
- Department of Medical Oncology, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris & Saint Cloud, France.,UVSQ, Paris-Saclay University, Saint Cloud, France
| | - Jean-Yves Pierga
- Department of Medical Oncology, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris & Saint Cloud, France. .,Paris Descartes University, Paris, France.
| |
Collapse
|
17
|
Inflammatory Breast Cancer: Diagnostic, Molecular and Therapeutic Considerations. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00337-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
18
|
Di Bonito M, Cantile M, Botti G. Pathological and molecular characteristics of inflammatory breast cancer. Transl Cancer Res 2019; 8:S449-S456. [PMID: 35117122 PMCID: PMC8798351 DOI: 10.21037/tcr.2019.03.24] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/18/2019] [Indexed: 11/11/2022]
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive form of breast cancer characterized by the presence of many dermal tumor emboli in the papillary and reticular dermis of the skin overlying the breast. IBC patients, compared to other breast cancer patients, have more frequently metastatic axillary lymph nodes. IBC is often high grade, negative for hormone receptors and presents with amplification of the HER2 gene. Invasive IBC is frequently of ductal phenotype, even if a specific histological distinction for these lesions has not been described. The pathogenesis and evolution of IBC are strongly dependent upon tumor microenvironment, characterized by several macrophages/monocytes and lymphocytes. The tumor and microenvironment cells are well molecularly characterized, showing the main contributor of inflammatory pathways in tumor biology of IBC. In addition, several molecular alterations are described in this tumor, such as mutations of ERBB2, KRAS, BRAF, EGFR, PIK3CA, PTEN, AKT1, and AKT3 genes that could suggest a therapeutic stratification of IBC patients with the combination of different biological target therapies.
Collapse
Affiliation(s)
- Maurizio Di Bonito
- Pathology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Monica Cantile
- Pathology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
19
|
Toxicity of locoregional radiotherapy in combination with bevacizumab in patients with non-metastatic breast cancer (TOLERAB): Final long-term evaluation. PLoS One 2019; 14:e0221816. [PMID: 31469859 PMCID: PMC6716668 DOI: 10.1371/journal.pone.0221816] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/15/2019] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Few data are available concerning the safety of bevacizumab (B) in combination with locoregional radiation therapy (RT). The objective of this study was to evaluate the 5-year late toxicity of concurrent B and RT in non-metastatic breast cancer. MATERIALS AND METHODS This multicentre prospective study included non-metastatic breast cancer patients enrolled in phase 3 clinical trials evaluating B with concurrent RT versus RT alone. All patients received neoadjuvant or adjuvant chemotherapy and normofractionated breast or chest wall RT, with or without regional lymph node RT. B was administered at an equivalent dose of 5 mg/kg once a week for 1 year. The safety profile was evaluated 1, 3 and 5 years after completion of radiotherapy. RESULTS A total of 64 patients were included between November 2007 and April 2010. Median follow-up was 60 months (12-73) and 5-year late toxicity data were available for 46 patients. The majority of tumours were triple-negative (68.8%), tumour size <2cm (41.3%) with negative nodal status (50.8%). Median total dose of B was 15,000mg and median duration was 11.2 months. No grade ≥3 toxicity was observed. Only 8 patients experienced grade 1-2 toxicities: n = 3 (6.5%) grade 1 lymphedema, n = 2 (4.3%) grade 1 pain, n = 1 (2.2%) grade 2 lymphedema, n = 1 (2.2%) grade 1 fibrosis. Five-year overall survival was 93.8%, disease-free survival was 89% and locoregional recurrence-free survival was 93.1%. CONCLUSION Concurrent B and locoregional RT are associated with acceptable 5-year toxicity in patients with non-metastatic breast cancer. No grade ≥3 toxicity was observed.
Collapse
|
20
|
Stem Cells Inhibition by Bevacizumab in Combination with Neoadjuvant Chemotherapy for Breast Cancer. J Clin Med 2019; 8:jcm8050612. [PMID: 31064127 PMCID: PMC6572380 DOI: 10.3390/jcm8050612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/22/2022] Open
Abstract
Preclinical works have suggested cytotoxic chemotherapies may increase the number of cancer stem cells (CSC) whereas angiogenesis inhibition may decrease CSC proliferation. We developed a proof of concept clinical trial to explore bevacizumab activity on breast CSC. Breast cancer patients requiring preoperative chemotherapy were included in this open-label, randomized, prospective, multicenter phase II trial. All received FEC-docetaxel combination, and patients randomized in the experimental arm received concomitant bevacizumab. The primary endpoint was to describe ALDH1 (Aldehyde dehydrogenase 1) positive tumor cells rate before treatment and after the fourth cycle. Secondary objectives included safety, pathological complete response (pCR) rate, disease-free survival (DFS), relapse-free survival (RFS), and overall survival (OS). Seventy-five patients were included. ALDH1+ cells rate increase was below the predefined 5% threshold in both arms for the 32 patients with two time points available. Grade 3 or 4 adverse events rates were similar in both arms. A non-significant increase in pCR was observed in the bevacizumab arm (42.6% vs. 18.2%, p = 0.06), but survival was not improved (OS: p = 0.89; DFS: p = 0.45; and RFS: p = 0.68). The increase of ALDH1+ tumor cells rate after bevacizumab-based chemotherapy was less than 5%. However, as similar results were observed with chemotherapy alone, bevacizumab impact on breast CSC cells cannot be confirmed.
Collapse
|
21
|
Influence of Biologic Subtype of Inflammatory Breast Cancer on Response to Neoadjuvant Therapy and Cancer Outcomes. Clin Breast Cancer 2018; 18:e501-e506. [DOI: 10.1016/j.clbc.2017.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/01/2017] [Accepted: 10/02/2017] [Indexed: 01/02/2023]
|
22
|
Lim B, Woodward WA, Wang X, Reuben JM, Ueno NT. Inflammatory breast cancer biology: the tumour microenvironment is key. Nat Rev Cancer 2018; 18:485-499. [PMID: 29703913 DOI: 10.1038/s41568-018-0010-y] [Citation(s) in RCA: 221] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive disease that accounts for ~2-4% of all breast cancers. However, despite its low incidence rate, IBC is responsible for 7-10% of breast cancer-related mortality in Western countries. Thus, the discovery of robust biological targets and the development of more effective therapeutics in IBC are crucial. Despite major international efforts to understand IBC biology, genomic studies have not led to the discovery of distinct biological mechanisms in IBC that can be translated into novel therapeutic strategies. In this Review, we discuss these molecular profiling efforts and highlight other important aspects of IBC biology. We present the intrinsic characteristics of IBC, including stemness, metastatic potential and hormone receptor positivity; the extrinsic features of the IBC tumour microenvironment (TME), including various constituent cell types; and lastly, the communication between these intrinsic and extrinsic components. We summarize the latest perspectives on the key biological features of IBC, with particular emphasis on the TME as an important contributor to the aggressive nature of IBC. On the basis of the current understanding of IBC, we hope to develop the next generation of translational studies, which will lead to much-needed survival improvements in patients with this deadly disease.
Collapse
Affiliation(s)
- Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Wendy A Woodward
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoping Wang
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M Reuben
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
23
|
Zare A, Petrova A, Agoumi M, Amstrong H, Bigras G, Tonkin K, Wine E, Baksh S. RIPK2: New Elements in Modulating Inflammatory Breast Cancer Pathogenesis. Cancers (Basel) 2018; 10:cancers10060184. [PMID: 29874851 PMCID: PMC6025367 DOI: 10.3390/cancers10060184] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/29/2018] [Accepted: 06/04/2018] [Indexed: 12/16/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive form of breast cancer that is associated with significantly high mortality. In spite of advances in IBC diagnoses, the prognosis is still poor compared to non-IBC. Due to the aggressive nature of the disease, we hypothesize that elevated levels of inflammatory mediators may drive tumorigenesis and metastasis in IBC patients. Utilizing IBC cell models and patient tumor samples, we can detect elevated NF-κB activity and hyperactivation of non-canonical drivers of NF-κB (nuclear factor kappaB)-directed inflammation such as tyrosine phosphorylated receptor-interacting protein kinase 2 (pY RIPK2), when compared to non-IBC cells or patients. Interestingly, elevated RIPK2 activity levels were present in a majority of pre-chemotherapy samples from IBC patients at the time of diagnosis to suggest that patients at diagnosis had molecular activation of NF-κB via RIPK2, a phenomenon we define as “molecular inflammation”. Surprisingly, chemotherapy did cause a significant increase in RIPK2 activity and thus molecular inflammation suggesting that chemotherapy does not resolve the molecular activation of NF-κB via RIPK2. This would impact on the metastatic potential of IBC cells. Indeed, we can demonstrate that RIPK2 activity correlated with advanced tumor, metastasis, and group stage as well as body mass index (BMI) to indicate that RIPK2 might be a useful prognostic marker for IBC and advanced stage breast cancer.
Collapse
Affiliation(s)
- Alaa Zare
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
| | - Alexandra Petrova
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
| | - Mehdi Agoumi
- Anatomic Pathologist at DynalifeDx, Diagnostic Laboratory Services; Department of Laboratory Medicine and Pathology, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2R3, Canada.
| | - Heather Amstrong
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
| | - Gilbert Bigras
- Cross Cancer Institute Department of Laboratory Medicine and Pathology, University of Alberta, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada.
| | - Katia Tonkin
- Division of Medical Oncology, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada.
| | - Eytan Wine
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
| | - Shairaz Baksh
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
- Division of Medical Oncology, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada.
- Division of Experimental Oncology, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada.
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB T6G 2R7, Canada.
- Women and Children's Health Research Institute, Edmonton Clinic Health Academy (ECHA), University of Alberta, 4-081 11405 87 Avenue NW Edmonton, AB T6G 1C9, Canada.
| |
Collapse
|
24
|
Stebbing J, Shah K, Lit LC, Gagliano T, Ditsiou A, Wang T, Wendler F, Simon T, Szabó KS, O'Hanlon T, Dean M, Roslani AC, Cheah SH, Lee SC, Giamas G. LMTK3 confers chemo-resistance in breast cancer. Oncogene 2018; 37:3113-3130. [PMID: 29540829 PMCID: PMC5992129 DOI: 10.1038/s41388-018-0197-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/12/2018] [Accepted: 02/18/2018] [Indexed: 12/31/2022]
Abstract
Lemur tyrosine kinase 3 (LMTK3) is an oncogenic kinase that is involved in different types of cancer (breast, lung, gastric, colorectal) and biological processes including proliferation, invasion, migration, chromatin remodeling as well as innate and acquired endocrine resistance. However, the role of LMTK3 in response to cytotoxic chemotherapy has not been investigated thus far. Using both 2D and 3D tissue culture models, we found that overexpression of LMTK3 decreased the sensitivity of breast cancer cell lines to cytotoxic (doxorubicin) treatment. In a mouse model we showed that ectopic overexpression of LMTK3 decreases the efficacy of doxorubicin in reducing tumor growth. Interestingly, breast cancer cells overexpressing LMTK3 delayed the generation of double strand breaks (DSBs) after exposure to doxorubicin, as measured by the formation of γH2AX foci. This effect was at least partly mediated by decreased activity of ataxia-telangiectasia mutated kinase (ATM) as indicated by its reduced phosphorylation levels. In addition, our RNA-seq analyses showed that doxorubicin differentially regulated the expression of over 700 genes depending on LMTK3 protein expression levels. Furthermore, these genes were found to promote DNA repair, cell viability and tumorigenesis processes / pathways in LMTK3-overexpressing MCF7 cells. In human cancers, immunohistochemistry staining of LMTK3 in pre- and post-chemotherapy breast tumor pairs from four separate clinical cohorts revealed a significant increase of LMTK3 following both doxorubicin and docetaxel based chemotherapy. In aggregate, our findings show for the first time a contribution of LMTK3 in cytotoxic drug resistance in breast cancer.
Collapse
Affiliation(s)
- Justin Stebbing
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
| | - Kalpit Shah
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Lei Cheng Lit
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Teresa Gagliano
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Angeliki Ditsiou
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Tingting Wang
- Cancer Science Institute of Singapore, Centre for Life Sciences, 28 Medical Drive, #02-15, Singapore, Singapore
| | - Franz Wendler
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Thomas Simon
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Krisztina Sára Szabó
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Timothy O'Hanlon
- Cancer Genomics Research Laboratory, National Cancer Institute, Division of Cancer Epidemiology and Genetics, Leidos Biomedical Research Inc., Bethesda, MD, 20892, USA
| | - Michael Dean
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - April Camilla Roslani
- Department of Surgery, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Swee Hung Cheah
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Soo-Chin Lee
- Cancer Science Institute of Singapore, Centre for Life Sciences, 28 Medical Drive, #02-15, Singapore, Singapore
| | - Georgios Giamas
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK.
| |
Collapse
|
25
|
Dautruche A, Belin L, Cottu P, Bontemps P, Lemanski C, de la Lande B, Baumann P, Missohou F, Lévy C, Peignaux K, Reynaud-Bougnoux A, Denis F, Gobillion A, Pernin V, Kirova Y. Evaluation at 3 years of concurrent bevacizumab and radiotherapy for breast cancer: Results of a prospective study. Cancer Radiother 2018; 22:222-228. [PMID: 29650388 DOI: 10.1016/j.canrad.2017.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/04/2017] [Indexed: 10/17/2022]
Abstract
PURPOSE To determine the 3 years late toxicity among patients with non-metastatic breast cancer who received concurrent bevacizumab and locoregional radiotherapy. MATERIAL AND METHODS This is a single-arm, multicentre, prospective study, of the toxicity of adjuvant concomitant association of bevacizumab and radiotherapy in patients with breast cancer. Toxicity was assessed by the Common Terminology Criteria for Adverse Events version 3.0 during the radiotherapy and follow-up clinics at 12 and 36 months after its completion. The study was designed to evaluate the toxicity at one year, 3 years and 5 years. RESULTS Sixty-four patients were included from October 2007 to August 2010. All of them received concurrent adjuvant radiotherapy and bevacizumab (in 24 cases after primary systemic treatment). All patients received non-fractionated radiotherapy to breast or chest wall with or without irradiation of regional lymph nodes. Early toxicity has been previously reported. Median follow-up was 46.4 months (range: 18-77 months). Median age was 53 years old (range: 23-68 years). The 3-years overall survival was 93% (range: 87-100%). Evaluation of the toxicity at 3 years was available for 67% of the patients. There was a low rate of toxicity: 14% grade 1 pain, 9% grade 1 fibrosis, 2% grade 1 telangiectasia, 2% grade 1 paresis, 7% grade 1 lymphedema and 2% grade 3 lymphedema. No grade 4 toxicity was observed. No patient had a left ventricular ejection fraction below 50% at 3 years. CONCLUSIONS Concurrent bevacizumab with locoregional radiotherapy is associated with acceptable 3-years toxicity in patients with breast cancer.
Collapse
Affiliation(s)
- A Dautruche
- Radiotherapy department, institut Curie, 26, rue d'Ulm, 75005 Paris, France.
| | - L Belin
- Biostatistics department, institut Curie, 26, rue d'Ulm, 75005 Paris, France
| | - P Cottu
- Oncology department, institut Curie, 26, rue d'Ulm, 75005 Paris, France
| | - P Bontemps
- Radiotherapy department, CHU Jean-Minjoz, 25030 Besançon, France
| | - C Lemanski
- Radiotherapy department, institut régional du cancer de Montpellier, 34298 Montpellier, France
| | - B de la Lande
- Radiotherapy department, institut Curie, René-Huguenin hospital, 92210 Saint-Cloud, France
| | - P Baumann
- Radiotherapy department, centre d'oncologie de Gentilly, 54000 Nancy, France
| | - F Missohou
- Radiotherapy department, centre Henri-Becquerel, 76038 Rouen, France
| | - C Lévy
- Radiotherapy department, centre François-Baclesse, 14000 Caen, France
| | - K Peignaux
- Radiotherapy department, centre Georges-François-Leclerc, 21079 Dijon, France
| | | | - F Denis
- Radiotherapy department, centre Jean-Bernard, 72000 Le Mans, France
| | - A Gobillion
- Biostatistics department, institut Curie, 26, rue d'Ulm, 75005 Paris, France
| | - V Pernin
- Radiotherapy department, institut Curie, 26, rue d'Ulm, 75005 Paris, France
| | - Y Kirova
- Radiotherapy department, institut Curie, 26, rue d'Ulm, 75005 Paris, France
| |
Collapse
|
26
|
Palazzo A, Dellapasqua S, Munzone E, Bagnardi V, Mazza M, Cancello G, Ghisini R, Iorfida M, Montagna E, Goldhirsch A, Colleoni M. Phase II Trial of Bevacizumab Plus Weekly Paclitaxel, Carboplatin, and Metronomic Cyclophosphamide With or Without Trastuzumab and Endocrine Therapy as Preoperative Treatment of Inflammatory Breast Cancer. Clin Breast Cancer 2018; 18:328-335. [PMID: 29486983 DOI: 10.1016/j.clbc.2018.01.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/23/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Inflammatory breast cancer (IBC) is a rare and highly aggressive disease. A neoadjuvant regimen with chemotherapy and an antiangiogenic strategy was investigated. PATIENTS AND METHODS Patients with primary or recurrent IBC who were candidates for neoadjuvant treatment received weekly carboplatin and paclitaxel plus bevacizumab every 3 weeks and oral metronomic cyclophosphamide for 6 months. Trastuzumab was added for patients with HER2+ tumors and endocrine therapy was added for patients with estrogen receptor and/or progesterone receptor ≥ 10% tumors. Oral metronomic capecitabine and cyclophosphamide was continued for 6 months after surgery in those patients with a response. The primary efficacy endpoints were pathologic complete remission (pCR) and the objective response. RESULTS From July 2010 to December 2013, 34 patients with IBC were included. The surrogate intrinsic tumor subtypes were as follows: luminal B-like (HER2-), 10 (29%); luminal B-like (HER2+), 8 (24%); HER2+ (nonluminal), 6 (18%); and triple negative, 10 (29%). An objective response was obtained in 30 patients (88%; 95% confidence interval, 73%-97%) and a pCR in 10 patients (29%; 95% confidence interval, 15%-48%). The proportion of pCR was significantly greater in the patients with HER2+ tumors (57%) than in patients with triple-negative (20%) or luminal B-like (HER2-) tumors (0%; P = .019). After a median follow-up of 4.4 years, the 5-year disease-free survival and overall survival was 58% and 72%, respectively. The achievement of pCR was associated with longer disease-free (P = .12) and overall (P = .029) survival. CONCLUSION In patients with IBC, neoadjuvant treatment with the investigated regimen was successful and well tolerated. Further studies evaluating the potential benefit of an antiangiogenic strategy in this setting are awaited.
Collapse
Affiliation(s)
- Antonella Palazzo
- Division of Medical Senology, European Institute of Oncology, Milan, Italy.
| | - Silvia Dellapasqua
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| | - Elisabetta Munzone
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| | - Vincenzo Bagnardi
- Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
| | - Manuelita Mazza
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| | - Giuseppe Cancello
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| | - Raffaella Ghisini
- Data Management, Clinical Trial Office, Scientific Direction, European Institute of Oncology, Milan, Italy
| | - Monica Iorfida
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| | - Emilia Montagna
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| | - Aaron Goldhirsch
- Scientific and Clinical Evaluation Board, European Institute of Oncology and International Breast Cancer Study Group, Milan, Italy
| | - Marco Colleoni
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| |
Collapse
|
27
|
Gampenrieder SP, Westphal T, Greil R. Antiangiogenic therapy in breast cancer. MEMO-MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2017; 10:194-201. [PMID: 29250196 PMCID: PMC5725520 DOI: 10.1007/s12254-017-0362-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 10/23/2017] [Indexed: 12/21/2022]
Abstract
Based on a strong rationale for anti-VEGF (vascular endothelial growth factor) treatment in breast cancer and promising preclinical data, great hopes have been placed on the anti-VEGF antibody bevacizumab. Clinical trials, however, reported conflicting results. In metastatic human epidermal growth factor receptor 2(HER2)-negative breast cancer, the addition of bevacizumab to standard chemotherapy improved consistently progression-free survival (PFS), however, without effect on overall survival (OS). In early breast cancer bevacizumab increased the pathologic complete response rate (pCR) after neoadjuvant therapy, but adjuvant trials did not demonstrate an effect on long-term survival. Unfortunately, despite extensive research, there is still no biomarker for bevacizumab efficacy available, making patient selection difficult. This review summarizes all phase III trials investigating efficacy and toxicity of bevacizumab in early, locally advanced and metastatic breast cancer. It recapitulates the main toxicities, gives an overview on biomarker studies and discusses the role and future aspects of antiangiogenic therapy in breast cancer.
Collapse
Affiliation(s)
- Simon Peter Gampenrieder
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria.,Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg Cancer Research Institute, Salzburg, Austria.,Cancer Cluster Salzburg, Salzburg, Austria
| | - Theresa Westphal
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria.,Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg Cancer Research Institute, Salzburg, Austria.,Cancer Cluster Salzburg, Salzburg, Austria
| | - Richard Greil
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria.,Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg Cancer Research Institute, Salzburg, Austria.,Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
28
|
Chinen LTD, Abdallah EA, Braun AC, Flores BDCTDCP, Corassa M, Sanches SM, Fanelli MF. Circulating Tumor Cells as Cancer Biomarkers in the Clinic. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 994:1-41. [PMID: 28560666 DOI: 10.1007/978-3-319-55947-6_1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is believed that the development of metastatic cancer requires the presence of circulating tumor cells (CTCs) , which are found in a patient's circulation as rare abnormal cells comingled with billions of the normal red and white blood cells. The systems developed for detection of CTCs have brought progress to cancer treatment. The molecular characterization of CTCs can aid in the development of new drugs, and their presence during treatment can help clinicians determine the prognosis of the patient. Studies have been carried out in patients early in the disease course, with only primary tumors, and the role of CTCs in prognosis seems to be as important as it is in patients with metastatic disease. The published studies on CTCs have focused on their prognostic significance, their utility in real-time monitoring of therapies, the identification of therapeutic and resistance targets, and understanding the process of metastasis . The analysis of CTCs during the early stages, as a "liquid biopsy," helps to monitor patients at different points in the disease course, including minimal residual disease, providing valuable information about the very early assessment of treatment effectiveness. Finally, CTCs can be used to screen patients with family histories of cancer or with diseases that can lead to the development of cancer. With standard protocols, this easily obtained and practical tool can be used to prevent the growth and spread of cancer. In this chapter, we review some important aspects of CTCs , surveying the disease aspects where these cells have been investigated.
Collapse
Affiliation(s)
| | - Emne Ali Abdallah
- International Research Center, A. C. Camargo Cancer Center, Rua Taguá 440, São Paulo, SP, 01508-010, Brazil
| | - Alexcia Camila Braun
- International Research Center, A. C. Camargo Cancer Center, Rua Taguá 440, São Paulo, SP, 01508-010, Brazil
| | | | - Marcelo Corassa
- Department of Medical Oncology, A. C. Camargo Cancer Center, Rua Professor Antônio Prudente, São Paulo, SP, 01509-010, Brazil
| | - Solange Moraes Sanches
- Department of Medical Oncology, A. C. Camargo Cancer Center, Rua Professor Antônio Prudente, São Paulo, SP, 01509-010, Brazil
| | - Marcello Ferretti Fanelli
- Department of Medical Oncology, A. C. Camargo Cancer Center, Rua Professor Antônio Prudente, São Paulo, SP, 01509-010, Brazil
| |
Collapse
|
29
|
Woodward WA, Cristofanilli M, Merajver SD, Van Laere S, Pusztai L, Bertucci F, Berditchevski F, Polyak K, Overmoyer B, Devi GR, Sterneck E, Schneider R, Debeb BG, Wang X, van Golen KL, El-Zein R, Rahal OM, Alexander A, Reuben JM, Krishnamurthy S, Lucci A, Ueno NT. Scientific Summary from the Morgan Welch MD Anderson Cancer Center Inflammatory Breast Cancer (IBC) Program 10 th Anniversary Conference. J Cancer 2017; 8:3607-3614. [PMID: 29667990 PMCID: PMC5687177 DOI: 10.7150/jca.21200] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/28/2017] [Indexed: 02/01/2023] Open
Abstract
In 2006, a remarkable collaboration between University of Texas MD Anderson Cancer Center clinicians and Texas and New Mexico State legislators led to the formation of a dedicated IBC Research Program and Clinic at MD Anderson. This initiative provided funding and infrastructure to foster coordination of an IBC World Consortium of national and international experts, and launch the first ever IBC international conference in 2008, which brought together experts from around the world to facilitate collaborations and accelerate progress. Indeed great progress has been made since then. National and international experts in IBC convened at the 10th Anniversary Conference of the MD Anderson IBC Clinic and Research Program and presented the most extensive sequencing analysis to date comparing IBC to non-IBC, gene- and protein-based immunoprofiling of IBC versus non-IBC patients, and converging lines of evidence on the specific role of the microenvironment in IBC. Novel models, unique metabolic mechanisms, and prominent survival pathways have been identified and were presented. Multiple clinical trials based on the work of the last decade are in progress or in development. The important challenges ahead were discussed. This progress and a coordinated summary of these works are presented herein.
Collapse
Affiliation(s)
- Wendy A Woodward
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Massimo Cristofanilli
- Developmental Therapeutics Program of Division of Hematology Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Sofia D Merajver
- Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI.,Department of Internal Medicine, University of Michigan, Ann Arbor, MI.,University of Michigan Comprehensive Cancer Center, Ann Arbor, MI, 48109, USA. The Office for Health Equity and Inclusion, University of Michigan, Ann Arbor, MI.,Program in Cancer Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Steven Van Laere
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp Belgium
| | - Lajos Pusztai
- Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Francois Bertucci
- Department of Medical Oncology, Institute Paoli-Calmettes, Marseille, France
| | - Fedor Berditchevski
- School of Cancer Sciences of the University of Birmingham, Birmingham, United Kingdom
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA.,Department of Medicine, Harvard Medical School, Boston, MA.,BBS Program, Harvard Medical School, Boston, MA.,Broad Institute, Cambridge, MA
| | - Beth Overmoyer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Gayathri R Devi
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Sciences, Durham, NC.,Women's Cancer Program, Duke Cancer Institute, Durham, NC
| | - Esta Sterneck
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, Frederick, MD
| | - Robert Schneider
- Department of Microbiology, New York University School of Medicine, New York, NY.,Perlmutter Cancer Center, New York University School of Medicine, New York, NY
| | - Bisrat G Debeb
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiaoping Wang
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kenneth L van Golen
- Department of Biological Sciences and The Center for Translational Cancer Research, The University of Delaware, Newark, DE
| | - Randa El-Zein
- Department of Radiology, Houston Methodist Research Institute, Houston, TX.,Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Omar M Rahal
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Angela Alexander
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - James M Reuben
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Savitri Krishnamurthy
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anthony Lucci
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naoto T Ueno
- MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
30
|
Costa R, Santa-Maria CA, Rossi G, Carneiro BA, Chae YK, Gradishar WJ, Giles FJ, Cristofanilli M. Developmental therapeutics for inflammatory breast cancer: Biology and translational directions. Oncotarget 2017; 8:12417-12432. [PMID: 27926493 PMCID: PMC5355355 DOI: 10.18632/oncotarget.13778] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/07/2016] [Indexed: 12/16/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive form of breast cancer, which accounts for approximately 3% of cases of breast malignancies. Diagnosis relies largely on its clinical presentation, and despite a characteristic phenotype, underlying molecular mechanisms are poorly understood. Unique clinical presentation indicates that IBC is a distinct clinical and biological entity when compared to non-IBC. Biological understanding of non-IBC has been extrapolated into IBC and targeted therapies for HER2 positive (HER2+) and hormonal receptor positive non-IBC led to improved patient outcomes in the recent years. This manuscript reviews recent discoveries related to the underlying biology of IBC, clinical progress to date and suggests rational approaches for investigational therapies.
Collapse
Affiliation(s)
- Ricardo Costa
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America
| | - Cesar A Santa-Maria
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Giovanna Rossi
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Benedito A Carneiro
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Young Kwang Chae
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - William J Gradishar
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Francis J Giles
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| | - Massimo Cristofanilli
- Developmental Therapeutics Program, Division of Hematology/Oncology, Feinberg School of Medicine, Chicago, United States of America.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, United States of America
| |
Collapse
|
31
|
Gonçalves A, Monneur A, Viens P, Bertucci F. The use of systemic therapies to prevent progression of inflammatory breast cancer: which targeted therapies to add on cytotoxic combinations? Expert Rev Anticancer Ther 2017; 17:593-606. [PMID: 28506194 DOI: 10.1080/14737140.2017.1330655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Inflammatory breast cancer is a rare but frequently fatal disease, essentially because of its high ability to develop distant metastases. Even though the prognosis of IBC was significantly improved by multimodal management, including the systematic use of cytotoxic-based induction, the prognosis remains largely dismal. Areas covered: This review presents the main achievements in the systemic treatment of IBC during the past 30 years. It focuses more specifically on recent results obtained with targeted therapies, including anti-HER2 and anti-angiogenic agents. Novel approaches under investigation are presented. Expert commentary: Current management of IBC is subtype-specific and the largest benefit has been achieved in HER2-positive disease. The identification of breakthrough therapeutic advances is eagerly awaited and will require the development of IBC-specific clinical trials. Future clinical investigations should not only aim to increase the pathological response rate but also to eradicate distant metastases, which ultimately lead to patient death.
Collapse
Affiliation(s)
- Anthony Gonçalves
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - Audrey Monneur
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - Patrice Viens
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - François Bertucci
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| |
Collapse
|
32
|
Challenging a Misnomer? The Role of Inflammatory Pathways in Inflammatory Breast Cancer. Mediators Inflamm 2017; 2017:4754827. [PMID: 28607534 PMCID: PMC5457777 DOI: 10.1155/2017/4754827] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/18/2017] [Indexed: 12/26/2022] Open
Abstract
Inflammatory breast cancer is a rare, yet highly aggressive form of breast cancer, which accounts for less than 5% of all locally advanced presentations. The clinical presentation of inflammatory breast cancer often differs significantly from that of noninflammatory breast cancer; however, immunohistochemistry reveals few, if any, distinguishing features. The more aggressive triple-negative and HER2-positive breast cancer subtypes are overrepresented in inflammatory breast cancer compared with noninflammatory breast cancer, with a poorer prognosis in response to conventional therapies. Despite its name, there remains some controversy regarding the role of inflammation in inflammatory breast cancer. This review summarises the current molecular evidence suggesting that inflammatory signaling pathways are upregulated in this disease, including NF-κB activation and excessive IL-6 production among others, which may provide an avenue for novel therapeutics. The role of the tumor microenvironment, through tumor-associated macrophages, infiltrating lymphocytes, and cancer stem cells is also discussed, suggesting that these tumor extrinsic factors may help account for the differences in behavior between inflammatory breast cancer and noninflammatory breast cancer. While there are various novel treatment strategies already underway in clinical trials, the need for further development of preclinical models of this rare but aggressive disease is paramount.
Collapse
|
33
|
Zambonin V, De Toma A, Carbognin L, Nortilli R, Fiorio E, Parolin V, Pilotto S, Cuppone F, Pellini F, Lombardi D, Pollini GP, Tortora G, Bria E. Clinical results of randomized trials and 'real-world' data exploring the impact of Bevacizumab for breast cancer: opportunities for clinical practice and perspectives for research. Expert Opin Biol Ther 2017; 17:497-506. [PMID: 28133971 DOI: 10.1080/14712598.2017.1289171] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Angiogenesis plays a fundamental role in breast cancer (BC) growth, progression and metastatic spread. After the promising introduction of bevacizumab for the treatment of advanced BC, the initial enthusiasm decreased when the FDA withdrew its approval in 2011. Nevertheless, several clinical studies exploring the role of bevacizumab have been subsequently published. Areas covered: The aim of this study is to review the available clinical trials exploring the potential effectiveness of bevacizumab in BC, regardless of the disease setting. Expert opinion: Even if the evidence suggests that bevacizumab must be ruled out from the HER2-positive and adjuvant setting, bevacizumab's benefit remains uncertain in the neoadjuvant setting and in the advanced treatment of HER2-negative patients. In the first setting, the addition of bevacizumab to chemotherapy increased the pathological complete response (pCR) rate in most clinical trials. However, the current absence of evidence that pCR is a trial-level surrogate for survival requires waiting for long-term results. In the advanced setting, all trials showed a benefit in progression-free survival, but not in overall survival, highlighting an increase of adverse events. The lack of predictors of response represents the main unmet need in which future clinical research will undoubtedly invest.
Collapse
Affiliation(s)
- Valentina Zambonin
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Alessandro De Toma
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Luisa Carbognin
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Rolando Nortilli
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Elena Fiorio
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Veronica Parolin
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Sara Pilotto
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | | | - Francesca Pellini
- b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,d Chirurgia Senologica, Azienda Ospedaliera Universitaria Integrata, A.O.U.I. Breast Surgery Verona , Verona , Italy
| | - Davide Lombardi
- b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,d Chirurgia Senologica, Azienda Ospedaliera Universitaria Integrata, A.O.U.I. Breast Surgery Verona , Verona , Italy
| | - Giovanni Paolo Pollini
- b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,d Chirurgia Senologica, Azienda Ospedaliera Universitaria Integrata, A.O.U.I. Breast Surgery Verona , Verona , Italy
| | - Giampaolo Tortora
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Emilio Bria
- a U.O. Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy.,b Breast Unit, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| |
Collapse
|
34
|
Effective personalized therapy for breast cancer based on predictions of cell signaling pathway activation from gene expression analysis. Oncogene 2017; 36:3553-3561. [PMID: 28135251 DOI: 10.1038/onc.2016.503] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 11/16/2016] [Accepted: 11/21/2016] [Indexed: 12/12/2022]
Abstract
Current therapeutic outcomes for breast cancer underscore the complexity of treating a heterogeneous disease. Indeed, studies have shown that differences in gene expression among patients with the same subtype of breast cancer are correlated with the response to treatment. This strongly suggests that there is an urgent need to treat breast cancer with a personalized approach. Here we employed cell signaling pathway signatures to predict pathway activity in subtypes of MMTV-Myc mammary tumors. We then split tumors into subsets and developed individualized combinatorial treatments for two subtypes with distinct pathway activation patterns. Elevation of the EGFR, RAS and TGFβ pathways was observed in one subtype whereas these pathways were not predicted to be active in the other subtype that had high predicted activity of the Myc, Stat3 and Akt pathways. In a proof-of-principle experiment, treatment of these two subtypes with targeted therapies inhibited tumor growth only in the subtype of tumor where the therapy was designed to be active. We then analyzed gene expression profiles of human breast cancer patients and patient-derived xenograft (PDX) samples to predict pathway activity, and validated our approach of developing individualized treatments in mice with PDX tumors. Importantly, our combinatorial therapy resulted in tumor regression, including regression in PDX samples from triple-negative breast cancer. Together our data is a proof-of-principle experiment that demonstrates that cell signaling pathway signature-guided treatment for breast cancer is viable.
Collapse
|
35
|
Pierga JY, Bidard FC, Autret A, Petit T, Andre F, Dalenc F, Levy C, Ferrero JM, Romieu G, Bonneterre J, Lerebours F, Bachelot T, Kerbrat P, Campone M, Eymard JC, Mouret-Reynier MA, Gligorov J, Hardy-Bessard AC, Lortholary A, Soulie P, Boher JM, Proudhon C, Charafe-Jaufret E, Lemonnier J, Bertucci F, Viens P. Circulating tumour cells and pathological complete response: independent prognostic factors in inflammatory breast cancer in a pooled analysis of two multicentre phase II trials (BEVERLY-1 and -2) of neoadjuvant chemotherapy combined with bevacizumab. Ann Oncol 2017; 28:103-109. [DOI: 10.1093/annonc/mdw535] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
36
|
Bertucci F, Goncalves A, Viens P. Bevacizumab in HER2-negative inflammatory breast cancer. Oncoscience 2016; 3:297-298. [PMID: 28105446 PMCID: PMC5235910 DOI: 10.18632/oncoscience.324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 11/08/2016] [Indexed: 01/05/2023] Open
Affiliation(s)
- François Bertucci
- Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Anthony Goncalves
- Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Patrice Viens
- Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| |
Collapse
|
37
|
Ma X, Wang X, Huang J, Chen Y, Zhang J, Zhang B, Shi C, Liu L. Bevacizumab Addition in Neoadjuvant Treatment Increases the Pathological Complete Response Rates in Patients with HER-2 Negative Breast Cancer Especially Triple Negative Breast Cancer: A Meta-Analysis. PLoS One 2016; 11:e0160148. [PMID: 27579484 PMCID: PMC5006981 DOI: 10.1371/journal.pone.0160148] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 07/14/2016] [Indexed: 02/05/2023] Open
Abstract
Background Neoadjuvant therapy is administered to breast cancer patients as an induction process before surgery or radiotherapy to reduce tumor size. Human epidermal growth factor receptor-2 (HER-2) negative breast cancer lacks effective standard target therapy. Bevacizumab has a controversial role in the treatment of breast cancer and we conduct a meta-analysis to evaluate the value of adding bevacizumab in neoadjuvant regimen. Methods Potentially eligible studies were retrieved using PubMed, EMBASE and Medline. Clinical characteristics of patients and statistical data with pathological complete response (pCR) data were collected. Then a meta-analysis model was established to investigate the correlation between administration of bevacizumab in neoadjuvant therapy and pCR rates in HER-2 negative breast cancer. Results Seven eligible studies and 5408 patients were yielded. The pCR rates for “breast” or “breast plus lymph node” were similar. In subgroup analysis, we emphasized on patients with triple-negative breast cancer (TNBC). In the criterion of “lesions in breast” the pooled ORs was 1.55 [1.29, 1.86], P<0.00001 and regarding to the evaluation criterion of “lesions in breast and lymph nodes”, the pooled ORs was 1.48 [1.23, 1.78], P<0.0001, in favor of bevacizumab administration. Conclusion According to our pooled results, we finally find that bevacizumab addition as a neoadjuvant chemotherapy component, for induction use with limited cycle to improve the pCR rates and patients may avoid long-term adverse event and long-term invalid survival improvement. Especially in subgroup analysis, pCR rates could be improved significantly and physicians could consider bevacizumab with caution. As patients could avoid the adverse event caused by long-term using of bevacizumab, long-term quality of life improvement may be achieved, especially in TNBC.
Collapse
Affiliation(s)
- Xuelei Ma
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoshan Wang
- Department of Clinical Oncology, Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Jingwen Huang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingtai Chen
- Department of Abdominal Surgery, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical Collage, Beijing, China
| | - Jing Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Binglan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Changle Shi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
38
|
Cristofanilli M. Inflammatory breast cancer: a new approach. Lancet Oncol 2016; 17:544-6. [PMID: 27032300 DOI: 10.1016/s1470-2045(16)00064-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 01/22/2016] [Indexed: 01/02/2023]
|