1
|
Lafci O, Resch D, Santonocito A, Clauser P, Helbich T, Baltzer PAT. Role of imaging based response assesment for adapting neoadjuvant systemic therapy for breast cancer: A systematic review. Eur J Radiol 2025; 187:112105. [PMID: 40252279 DOI: 10.1016/j.ejrad.2025.112105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
PURPOSE The objective of this systematic review is to investigate the role of imaging in response monitoring during neoadjuvant systemic therapy (NST) for breast cancer and assess whether treatment modifications based on imaging response are implemented in clinical practice. METHODS A systematic review was conducted, analyzing five clinical practice guidelines and 147 clinical trial publications involving NST for breast cancer. The snowballing technique was employed, using a "start set" of clinical guidelines to trace relevant trials. Additionally, a PubMed search was conducted to identify trials published between 2023-2024. The review analyzed the use of imaging modalities, timing, and response criteria, and whether escalation, de-escalation, or change of treatment occurred based on imaging response. RESULTS Imaging was utilized in 81 % (119/147) of the trials, with ultrasound, MRI, and mammography being the most frequently employed modalities. Mid-treatment imaging was applied in 56 % (83/147) of the trials. However, only 15 % (22/147) of the trials implemented treatment modifications based on imaging response, highlighting the limited application of imaging response-guided therapy. No standardized imaging protocols or consistent response-guided treatment strategies were identified across the trials or clinical practice guidelines, with considerable variability in imaging methods, timing, and response criteria. CONCLUSION This systematic review underscores the critical need for standardized imaging protocols, response assessment criteria and image-guided treatment decisions. It is therefore evident that imaging for response monitoring during treatment should preferably be performed within clinical trials.
Collapse
Affiliation(s)
- Oguz Lafci
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Daphne Resch
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Ambra Santonocito
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Paola Clauser
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Thomas Helbich
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Pascal A T Baltzer
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
2
|
Li Z, Guo S, Xue H, Li L, Guo Y, Duan S, Zhu H. Efficacy and safety of trastuzumab deruxtecan in the treatment of HER2-low/positive advanced breast cancer: a single-arm meta-analysis. Front Pharmacol 2023; 14:1183514. [PMID: 37426807 PMCID: PMC10324614 DOI: 10.3389/fphar.2023.1183514] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Background: Clinical trials have shown that the use of trastuzumab deruxtecan (DS-8201) alone is expected to provide novel therapeutic options for HER2-low/positive patients. Nevertheless, there are some variations in the efficacy of trial results, with potential risks at the safety level. Most DS-8201 trials in HER2 advanced breast cancer (ABC) have been conducted in the form of small-sample nonrandomized controlled studies, resulting in a lack of validated indicators to evaluate the efficacy and safety of DS-8201. Thus, this meta-analysis aimed to pool the results of various trials of DS-8201 alone to explore the efficacy and safety of DS-8201 in patients with HER2-low/positive advanced breast cancer. Methods: Relevant studies were searched in seven databases, including Embase, PubMed, Web of Science, Cochrane Library, CNKI, VIP database and WanFang data, to collect single-arm studies on DS-8201 for HER2-low/positive ABC. MINORS was adopted for quality assessment and STATA 16.0 for data analysis. Results: Ten studies involving 1,108 patients were included in this meta-analysis. As for the tumor response rate, the pooled ORR and DCR of all studies reached 57% (95% CI: 47%-67%) and 92% (95% CI: 89%-96%) respectively, and the pooled ORRs of the HER2-low expression group and the HER2-positive expression group were 46% (95% CI: 35%-56%) and 64% (95% CI: 54%-74%). Only the low expression group achieved median survival time, with a pooled median PFS and median OS of 9.24 (95% CI: 7.54-10.94) months and 23.87 (95% CI: 21.56-26.17) months, respectively. The most common treatment-related adverse events from DS-8201 were nausea (all grades: 62%; ≥ grade III: 5%), fatigue (all grade: 44%; ≥ grade III: 6%), and alopecia (all grades: 38%; ≥ grade III: 0.5%). Drug-related interstitial lung disease or pneumonitis occurred in 13% of the 1,108 patients, with only a 1% incidence of AE ≥ grade III. Conclusion: The present study suggests that DS-8201 is effective and safe in the treatment of ABC with low or positive HER2 expression, providing additional relevant information for its clinical application. However, further strengthening of the pairs is needed, as well as more clinical studies to support individualized treatment. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42023390316.
Collapse
Affiliation(s)
- Zongyu Li
- Clinical Medical Research Institute, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - Shangwen Guo
- Clinical Medical Research Institute, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - Haoyi Xue
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - Luying Li
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - Yuyuan Guo
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - Sinuo Duan
- School of Medicine, Shihezi University of China, Shihezi Xinjiang Production and Construction Corps, Shihezi, China
| | - He Zhu
- Clinical Medical Research Institute, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
3
|
Zhou X, Lian H, Li H, Fan M, Xu W, Jin Y. Nanotechnology in cervical cancer immunotherapy: Therapeutic vaccines and adoptive cell therapy. Front Pharmacol 2022; 13:1065793. [PMID: 36588709 PMCID: PMC9802678 DOI: 10.3389/fphar.2022.1065793] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy is an emerging method for the treatment of cervical cancer and is more effective than surgery and radiotherapy, especially for recurrent cervical cancer. However, immunotherapy is limited by adverse effects in clinical practice. In recent years, nanotechnology has been widely used for tumor diagnosis, drug delivery, and targeted therapy. In the setting of cervical cancer, nanotechnology can be used to actively or passively target immunotherapeutic agents to tumor sites, thereby enhancing local drug delivery, reducing drug adverse effects, achieving immunomodulation, improving the tumor immune microenvironment, and optimizing treatment efficacy. In this review, we highlight the current status of therapeutic vaccines and adoptive cell therapy in cervical cancer immunotherapy, as well as the application of lipid carriers, polymeric nanoparticles, inorganic nanoparticles, and exosomes in this context.
Collapse
Affiliation(s)
- Xuyan Zhou
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Haiying Lian
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Hongpeng Li
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Meiling Fan
- Gynecology Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China,*Correspondence: Meiling Fan, ; Wei Xu, ; Ye Jin,
| | - Wei Xu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,*Correspondence: Meiling Fan, ; Wei Xu, ; Ye Jin,
| | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,*Correspondence: Meiling Fan, ; Wei Xu, ; Ye Jin,
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Moreira R, Granja A, Pinheiro M, Reis S. Nanomedicine Interventions in Clinical Trials for the Treatment of Metastatic Breast Cancer. APPLIED SCIENCES 2021; 11:1624. [DOI: 10.3390/app11041624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Abstract
Breast cancer was responsible for the deaths of 626,679 women in 2018. After decades of research, the mortality rates remain high. While the barrier of selectively killing tumor cells is not yet overcome, the search for targeted therapeutics continues. The use of nanomedicine in cancer treatment has opened up new possibilities for more precise drug-delivery systems. This review aimed to gather information and analyze recent clinical trials evaluating the therapeutic effects of nanoparticles in the treatment of metastatic breast cancer. To accomplish this, the clinicaltrials.gov database was consulted, and after employing specific exclusion criteria, 11 clinical trials were selected. Nanoparticle albumin-stabilized paclitaxel was evaluated in ten clinical trials and paclitaxel-incorporating polymeric micelles were assessed in one clinical trial. Overall, this review confirmed a clinical benefit in the use of nanoparticle albumin-stabilized paclitaxel for the treatment of breast cancer, with reduced toxicity when compared to first-line treatments. Three studies did not meet the primary endpoint, however, and so the authors advised further evaluations. Although the use of nanomedicine is revolutionizing the cancer field, to integrate this regimen into generalized clinical treatment, additional clinical trials must be performed to achieve a favorable safety and efficacy profile.
Collapse
Affiliation(s)
- Rita Moreira
- Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- ABC-RI, Algarve Biomedical Center Research Institute, 8005-139 Faro, Portugal
| | - Andreia Granja
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Marina Pinheiro
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Salette Reis
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
7
|
Yang X, Wu D, Yuan S. Tyrosine Kinase Inhibitors in the Combination Therapy of HER2 Positive Breast Cancer. Technol Cancer Res Treat 2020; 19:1533033820962140. [PMID: 33034269 PMCID: PMC7592330 DOI: 10.1177/1533033820962140] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2)-positive breast cancer (BC)
accounts for about 20% to 30% of all BC subtypes and is characterized by
invasive disease and poor prognosis. With the emergence of anti-HER2 target
drugs, HER2-positive BC patient outcomes have changed dramatically. However,
treatment failure is mostly due to drug resistance and the special treatment
needs of different subgroups. Small molecule tyrosine kinase inhibitors can
inhibit multiple targets of the human epidermal growth factor receptor family
and activate PI3K/AKT, MAPK, PLC γ, ERK1/2, JAK/STAT, and other pathways
affecting the expression of MDM2, mTOR, p27, and other transcription factors.
This can help regulate the differentiation, apoptosis, migration, growth, and
adhesion of normal cells and reverse drug resistance to a certain extent. These
inhibitors can cross the blood-brain barrier and be administered orally. They
have a good synergistic effect with effective drugs such as trastuzumab,
pertuzumab, t-dm1, and cyclin-dependent kinase 4 and 6 inhibitors. These
advantages have resulted in small-molecule tyrosine kinase inhibitors attracting
attention. The new small-molecule tyrosine kinase inhibitor was investigated in
multi-target anti-HER2 therapy, showed a good effect in preclinical and clinical
trials, and to some extent, improved the prognosis of HER2-positive BC patients.
Its use could lead to a de-escalation of treatment in some patients, possibly
preventing unnecessary procedures along with the associated side effects and
costs.
Collapse
Affiliation(s)
- Xue Yang
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - Dapeng Wu
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - Shengli Yuan
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|
8
|
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
|
9
|
Ji X, Jin C, Dong X, Dixon MS, Williams KP, Zheng W. Literature-Wide Association Studies (LWAS) for a Rare Disease: Drug Repurposing for Inflammatory Breast Cancer. Molecules 2020; 25:molecules25173933. [PMID: 32872166 PMCID: PMC7504746 DOI: 10.3390/molecules25173933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 01/04/2023] Open
Abstract
Drug repurposing is an effective means for rapid drug discovery. The aim of this study was to develop and validate a computational methodology based on Literature-Wide Association Studies (LWAS) of PubMed to repurpose existing drugs for a rare inflammatory breast cancer (IBC). We have developed a methodology that conducted LWAS based on the text mining technology Word2Vec. 3.80 million "cancer"-related PubMed abstracts were processed as the corpus for Word2Vec to derive vector representation of biological concepts. These vectors for drugs and diseases served as the foundation for creating similarity maps of drugs and diseases, respectively, which were then employed to find potential therapy for IBC. Three hundred and thirty-six (336) known drugs and three hundred and seventy (370) diseases were expressed as vectors in this study. Nine hundred and seventy (970) previously known drug-disease association pairs among these drugs and diseases were used as the reference set. Based on the hypothesis that similar drugs can be used against similar diseases, we have identified 18 diseases similar to IBC, with 24 corresponding known drugs proposed to be the repurposing therapy for IBC. The literature search confirmed most known drugs tested for IBC, with four of them being novel candidates. We conclude that LWAS based on the Word2Vec technology is a novel approach to drug repurposing especially useful for rare diseases.
Collapse
Affiliation(s)
| | | | | | | | - Kevin P. Williams
- Correspondence: (K.P.W.); (W.Z.); Tel.: +1-(919)-530-7726 (K.P.W.); +1-(919)-530-6752 (W.Z.)
| | - Weifan Zheng
- Correspondence: (K.P.W.); (W.Z.); Tel.: +1-(919)-530-7726 (K.P.W.); +1-(919)-530-6752 (W.Z.)
| |
Collapse
|
10
|
Martin Huertas R, Fernández Abad M, Corral de la Fuente E, Serrano Domingo JJ, Martínez Jáñez N. Early relapse of inflammatory breast carcinoma treated with lapatinib and capecitabine: Ten years of complete response. Breast J 2020; 26:800-802. [PMID: 31565830 PMCID: PMC7216947 DOI: 10.1111/tbj.13606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 11/30/2022]
|
11
|
Abo-Zeid MAM, Abo-Elfadl MT, Gamal-Eldeen AM. Evaluation of lapatinib cytotoxicity and genotoxicity on MDA-MB-231 breast cancer cell line. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 71:103207. [PMID: 31234033 DOI: 10.1016/j.etap.2019.103207] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 05/20/2019] [Accepted: 06/06/2019] [Indexed: 06/09/2023]
Abstract
Lapatinib, one of the tyrosine kinase inhibitors (TKIs), is used to reduce epidermal growth factor family proteins overexpression. This study aims to assess the cytotoxic and genotoxic effects of lapatinib on the triple negative breast cancer cell line "MDA-MB-231". We investigated the cytotoxicity of lapatinib by MTT assay, mode of cell death using apoptosis-necrosis assay, DNA damage using micronucleus test, EGFR protein expression by immunocytochemistry, and assessed its effect on EGFR (7p11.2 locus) and TP53 (17p13 locus) genes using interphase-FISH technique. Lapatinib induced cytotoxicity on MDA-MB-231 cell line by elevating the concentration and its IC50 value was 32.5 μM after 24 h. Lapatinib increased apoptotic cells and micronuclei in binucleated cells gradually by increasing the concentration for 24 h. The EGFR protein expression was reduced by double fold that expressed in non-treated cells. Lapatinib enhanced deletion of EGFR gene signals highly significantly from the lowest concentration. Alternatively, lapatinib amplified signals of TP53 gene effectively by raising the concentration. In conclusion, lapatinib induced cytotoxic and genotoxic effects on MDA-MB-231 cell line. However, laptinib reduced the EGFR protein expression and EGFR signals, it raised the apoptotic cells and TP53 gene signals, which triggered extensive DNA damage. Therefore, lapatinib is an effective TKI in triple negative breast cancer cells as elucidated by its mode of cell death.
Collapse
Affiliation(s)
- Mona A M Abo-Zeid
- Genetics and Cytology Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki, 12622, Cairo, Egypt; Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki, 12622, Cairo, Egypt.
| | - Mahmoud T Abo-Elfadl
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki, 12622, Cairo, Egypt; Biochemistry Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Amira M Gamal-Eldeen
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki, 12622, Cairo, Egypt; Biochemistry Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki, 12622, Cairo, Egypt; Clinical Laboratory Department, College of Applied Medical Sciences, Taif University, Al Mutamarat Rd, Al Mathnah, At Taif, 26521, Saudi Arabia
| |
Collapse
|
12
|
Inflammatory Breast Cancer. Breast Cancer 2019. [DOI: 10.1007/978-3-319-96947-3_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
13
|
Design, synthesis and preclinical evaluation of 5-methyl-N4-aryl-furo[2,3-d]pyrimidines as single agents with combination chemotherapy potential. Bioorg Med Chem Lett 2018; 28:3085-3093. [DOI: 10.1016/j.bmcl.2018.07.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/19/2018] [Accepted: 07/26/2018] [Indexed: 01/16/2023]
|
14
|
Rafnsdóttir SL, Audisio RA. Inflammatory Breast Cancer: What surgeons need to know. Eur J Surg Oncol 2018; 44:1139-1141. [PMID: 29954638 DOI: 10.1016/j.ejso.2018.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/06/2018] [Indexed: 01/09/2023] Open
Abstract
Strict criteria to identify Inflammatory Breast Cancer (IBC) have been made available - these are based on pathological as well as clinical observations. It is mandatory to confirm the criteria used in any further report. Scientific evidence accumulated so far is confusing, and unclear therefore no hard conclusion can be drawn from the available literature on the management of IBC. The advent of new medications results into an up-to-date management and different outcomes. It is mandatory to follow these criteria if reliable guidelines are to be made available in the next future.
Collapse
Affiliation(s)
- Svanheiður Lóa Rafnsdóttir
- Oncoplastic Breast Surgery, Department of Surgery, Landspitali University Hospital, 13-A Hringbraut, IS-101, Reykjavik, Iceland; Department of Surgery, Institute of Clinical Sciences, Sahlgrenska University Hospital, Blå Stråket 5, 413 45, Göteborg, Sweden.
| | - Riccardo A Audisio
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska University Hospital, Blå Stråket 5, 413 45, Göteborg, Sweden.
| |
Collapse
|
15
|
Curigliano G. Inflammatory breast cancer and chest wall disease: The oncologist perspective. Eur J Surg Oncol 2018; 44:1142-1147. [PMID: 30032791 DOI: 10.1016/j.ejso.2018.05.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/11/2018] [Indexed: 12/13/2022] Open
Abstract
Chest wall inflammatory and lymphangitic breast cancer represents a clinical spectrum and a model disease. Inflammation and the immune response have a role in the natural history of this special clinical presentation. Preclinical models and biomarker studies suggest that inflammatory breast cancer comprises a more important role for the tumour microenvironment, including immune cell infiltration and vasculogenesis, especially lympho-angiogenesis. Across this clinical continuum of the chest wall disease there is an important role of the inflammation cascade. The activation of mature dendritic cells (DCs) through toll like receptors (TLRs) or by inflammatory cytokines converts immature DCs into mature DCs that present specific antigen to T cells, thereby activating them. Maturation of DCs is accompanied by co-stimulatory molecules and secretion of inflammatory cytokines polarizing lymphocytic, macrophages and fibroblast infiltration. It is unknown whether immune cells associated to the IBC microenvironment play a role in this scenario to transiently promote epithelial to mesenchymal transition (EMT) in these cells. Immune and microenvirnment factors can induce phenotypic, morphological, and functional changes in breast cancer cells. We can hypothesize that similar inflammatory conditions in vivo may support both the rapid metastasis and tight tumor emboli that are characteristic of chest wall disease and that targeted anti-inflammatory therapy may play a role in this patient population. The current review will review biological and clinical data of this special condition.
Collapse
Affiliation(s)
- Giuseppe Curigliano
- University of Milano, Department of Oncology and Hemato-Oncology, Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milano, Italy.
| |
Collapse
|
16
|
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
|
17
|
Rosso KJ, Ueno NT, Woodward WA, Lucci A. In response to “outcomes of patients with inflammatory breast cancer treated by breast conserving surgery”: the argument against breast conservation and sentinel lymph node biopsy in IBC. Breast Cancer Res Treat 2017; 165:779-781. [DOI: 10.1007/s10549-017-4337-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/08/2017] [Indexed: 11/28/2022]
|
18
|
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
|
19
|
Curigliano G. Chest Wall Disease: The Clinical Continuum Between Inflammatory and Lymphangitic Breast Cancer. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Dawood S, Cristofanilli M. IBC as a Rapidly Spreading Systemic Disease: Clinical and Targeted Approaches Using the Neoadjuvant Model. J Natl Cancer Inst Monogr 2016; 2015:56-9. [PMID: 26063888 DOI: 10.1093/jncimonographs/lgv017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive form of invasive breast cancer accounting for 2.5% of all breast cancer cases. It is characterized by rapid progression, younger age of onset as compared with other cancers, local and distant metastases, and lower overall survival. The multidisciplinary management of IBC includes neoadjuvant systemic chemotherapy, surgery, radiotherapy, and hormonal therapy in hormone receptor-positive disease. Pathological complete response represents an important prognostic factor suggesting IBC as the ideal in-vivo model for therapeutic development. Molecular subtyping demonstrated higher frequency of basal-like an HER2 disease in IBC compared with non-IBC indicating the areas of novel therapeutic interventions. The prospective testing of HER2-targeted therapies (eg, trastuzumab and lapatinib) demonstrated the validity of this concept and the potential to change the outcome of this aggressive disease.
Collapse
Affiliation(s)
- Shaheenah Dawood
- Department of Medical Oncology, Dubai Hospital, United Arab Emirates (SD); Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA (MC)
| | - Massimo Cristofanilli
- Department of Medical Oncology, Dubai Hospital, United Arab Emirates (SD); Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA (MC).
| |
Collapse
|
21
|
Xu MM, Pu Y, Zhang Y, Fu YX. The Role of Adaptive Immunity in the Efficacy of Targeted Cancer Therapies. Trends Immunol 2016; 37:141-153. [PMID: 26778079 DOI: 10.1016/j.it.2015.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 12/10/2015] [Accepted: 12/13/2015] [Indexed: 01/22/2023]
Abstract
Accumulating evidence indicates that the efficacy of tumor-targeted therapies relies on the host immune response, including targeted small-molecule and antibody approaches that were not previously thought to have an immune component. Here, we review the current understanding of how targeted therapies on tumor cells could have a major impact on the immune response, and how this relates to the therapeutic efficacy of these approaches. In this context, we evaluate different strategies that combine targeted therapies with immunotherapy approaches, and discuss past and ongoing clinical trials. We highlight gaps in knowledge, and argue that significant progress for combined therapies will require a better understanding of the complex interactions between immune cells, the tumor, and the tumor microenvironment (TME) in different cancer settings.
Collapse
Affiliation(s)
- Meng Michelle Xu
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Yang Pu
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Yuan Zhang
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Yang-Xin Fu
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, IL 60637, USA; Department of Pathology and Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA.
| |
Collapse
|
22
|
Jhaveri K, Teplinsky E, Silvera D, Valeta-Magara A, Arju R, Giashuddin S, Sarfraz Y, Alexander M, Darvishian F, Levine PH, Hashmi S, Zolfaghari L, Hoffman HJ, Singh B, Goldberg JD, Hochman T, Formenti S, Esteva FJ, Moran MS, Schneider RJ. Hyperactivated mTOR and JAK2/STAT3 Pathways: Molecular Drivers and Potential Therapeutic Targets of Inflammatory and Invasive Ductal Breast Cancers After Neoadjuvant Chemotherapy. Clin Breast Cancer 2015; 16:113-22.e1. [PMID: 26774497 DOI: 10.1016/j.clbc.2015.11.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 11/09/2015] [Accepted: 11/12/2015] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Inflammatory breast cancer (IBC) is an aggressive and rare cancer with a poor prognosis and a need for novel targeted therapeutic strategies. Preclinical IBC data showed strong activation of the phosphatidylinositide-3-kinase/mammalian target of rapamycin (mTOR) and Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathways, and expression of inflammatory cytokines and tumor-associated macrophages (TAMs). PATIENTS AND METHODS Archival tumor tissue from 3 disease types (IBC treated with neoadjuvant chemotherapy [NAC], n = 45; invasive ductal carcinoma [IDC] treated with NAC [n = 24; 'treated IDC'; and untreated IDC [n = 27; 'untreated IDC']) was analyzed for the expression of biomarkers phospho-S6 (pS6) (mTOR), phospho-JAK2 (pJAK2), pSTAT3, interleukin (IL)-6, CD68 (monocytes, macrophages), and CD163 (TAMs). Surrounding nontumor tissue was also analyzed. RESULTS Biomarker levels and surrogate activity according to site-specific phosphorylation were shown in the tumor tissue of all 3 disease types but were greatest in IBC and treated IDC and least in untreated IDC for pS6, pJAK2, pSTAT3, and IL-6. Of 37 IBC patients with complete biomarker data available, 100% were pS6-positive and 95% were pJAK2-positive. In nontumor tissue, biomarker levels were observed in all groups but were generally greatest in untreated IDC and least in IBC, except for JAK2. CONCLUSION IBC and treated IDC display similar levels of mTOR and JAK2 biomarker activation, which suggests a potential mechanism of resistance after NAC. Biomarker levels in surrounding nontumor tissue suggested that the stroma might be activated by chemotherapy and resembles the oncogenic tumor-promoting environment. Activation of pS6 and pJAK2 in IBC might support dual targeting of the mTOR and JAK/STAT pathways, and the need for prospective studies to investigate combined targeted therapies in IBC.
Collapse
Affiliation(s)
- Komal Jhaveri
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY.
| | - Eleonora Teplinsky
- Division of Hematology & Medical Oncology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Deborah Silvera
- Department of Microbiology, New York University School of Medicine, New York, NY
| | - Amanda Valeta-Magara
- Department of Microbiology, New York University School of Medicine, New York, NY
| | - Rezina Arju
- Department of Microbiology, New York University School of Medicine, New York, NY
| | - Shah Giashuddin
- Department of Pathology, The Brooklyn Hospital Center, Brooklyn, NY
| | - Yasmeen Sarfraz
- Department of Microbiology, New York University School of Medicine, New York, NY
| | | | | | - Paul H Levine
- Department of Epidemiology and Biostatistics, George Washington University, Washington, DC
| | - Salman Hashmi
- Division of Biostatistics, New York University School of Medicine, New York, NY
| | - Ladan Zolfaghari
- Division of Biostatistics, New York University School of Medicine, New York, NY
| | - Heather J Hoffman
- Division of Biostatistics, New York University School of Medicine, New York, NY
| | - Baljit Singh
- Department of Epidemiology and Biostatistics, George Washington University, Washington, DC
| | - Judith D Goldberg
- Division of Biostatistics, New York University School of Medicine, New York, NY
| | - Tsivia Hochman
- Division of Biostatistics, New York University School of Medicine, New York, NY
| | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY
| | - Francisco J Esteva
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY
| | - Meena S Moran
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT
| | - Robert J Schneider
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY; Department of Microbiology, New York University School of Medicine, New York, NY
| |
Collapse
|
23
|
Abstract
The poor prognosis of inflammatory breast cancer (IBC) is due to its strong metastatic potential. During the last three decades, the introduction of neoadjuvant chemotherapy (CT), and its improvement with successive additions of anthracyclines and then taxanes, allowed to double the survival. However, the 5-year survival still remains lower than 50%, with the pathological complete response (pCR) to neoadjuvant CT being a major prognostic factor. Since 1995, several innovative approaches have been evaluated. Initially, the trials of high-dose CT with hematopoietic stem cell transplantation have generated promising results, but ultimately failed to change standards of treatment, in particular because of its toxicity. More recently, a few targeted therapies, combined to conventional CT, have been assessed, due to the frequent overexpression of HER2 and EGFR and the important vascularization of IBC. Trastuzumab, a monoclonal antibody targeting HER2, has shown a clear advantage in terms of pCR and survival in studies dedicated to, HER2-positive locally advanced breast cancers, including IBC. Lapatinib, a dual tyrosine kinase inhibitor anti-HER2 and EGFR, has shown significant activity in two phase II studies dedicated to HER2-positive IBC. The interest of HER2-double blockade by the combination of trastuzumab-pertuzumab combined to docetaxel has been demonstrated in term of pCR in the NEOSPHERE study which also included HER2-positive IBC. Among the anti-angiogenic drugs tested in studies dedicated to IBC, bevacizumab has given the most interesting results in term of efficacy/toxicity ratio. In the Beverly 2 study HER2-positive IBC patients were treated by the combination chemotherapy, trastuzumab and bevacizumab: the rate of pCR was 64%, and the 3-year disease-free and overall survivals were 68% and 90%, respectively; the increase of endothelial cells circulating was inversely correlated to the probability of pCR. All those treatments have been extrapolated from standard breast cancers. Thus, a deep molecular knowledge of IBC appears to be critical in order to develop specific treatments effectively targeting its particular aggressiveness.
Collapse
|
24
|
Identifying the impact of inflammatory breast cancer on survival: a retrospective multi-center cohort study. Arch Gynecol Obstet 2015; 292:655-64. [DOI: 10.1007/s00404-015-3691-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 03/13/2015] [Indexed: 01/08/2023]
|
25
|
|
26
|
van Uden DJP, van Laarhoven HWM, Westenberg AH, de Wilt JHW, Blanken-Peeters CFJM. Inflammatory breast cancer: an overview. Crit Rev Oncol Hematol 2014; 93:116-26. [PMID: 25459672 DOI: 10.1016/j.critrevonc.2014.09.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 08/08/2014] [Accepted: 09/30/2014] [Indexed: 12/14/2022] Open
Abstract
Inflammatory breast cancer (IBC) is the most aggressive entity of breast cancer. Management involves coordination of multidisciplinary management and usually includes neoadjuvant chemotherapy, ablative surgery if a tumor-free resection margin is expected and locoregional radiotherapy. This multimodal therapeutic approach has significantly improved patient survival. However, the median overall survival among women with IBC is still poor. By elucidating the biologic characteristics of IBC, new treatment options may become available. We performed a comprehensive review of the English-language literature on IBC through computerized literature searches. The objective of the current review is to present an overview of the literature related to the biology, imaging and multidisciplinary treatment of inflammatory breast cancer.
Collapse
Affiliation(s)
- D J P van Uden
- Department of Surgery, Canisius Wilhelmina Hospital, Postbus 9015, 6500 GS Nijmegen, The Netherlands.
| | - H W M van Laarhoven
- Medical Oncology, Academic Medical Center, University of Amsterdam, Postbus 22660, 1100 DD Amsterdam, The Netherlands
| | - A H Westenberg
- Institute for Radiation Oncology, Postbus 60160, 6800 JD Arnhem, The Netherlands
| | - J H W de Wilt
- Department of Surgery, Radboud University Medical Center, Geert Grooteplein-Zuid 10, 6525 GA Nijmegen, The Netherlands
| | | |
Collapse
|
27
|
Zhang X, Raghavan S, Ihnat M, Thorpe JE, Disch BC, Bastian A, Bailey-Downs LC, Dybdal-Hargreaves NF, Rohena CC, Hamel E, Mooberry SL, Gangjee A. The design and discovery of water soluble 4-substituted-2,6-dimethylfuro[2,3-d]pyrimidines as multitargeted receptor tyrosine kinase inhibitors and microtubule targeting antitumor agents. Bioorg Med Chem 2014; 22:3753-72. [PMID: 24890652 PMCID: PMC4089508 DOI: 10.1016/j.bmc.2014.04.049] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/16/2014] [Accepted: 04/25/2014] [Indexed: 12/13/2022]
Abstract
The design, synthesis and biological evaluations of fourteen 4-substituted 2,6-dimethylfuro[2,3-d]pyrimidines are reported. Four compounds (11-13, 15) inhibit vascular endothelial growth factor receptor-2 (VEGFR-2), platelet-derived growth factor receptor β (PDGFR-β), and target tubulin leading to cytotoxicity. Compound 11 has nanomolar potency, comparable to sunitinib and semaxinib, against tumor cell lines overexpressing VEGFR-2 and PDGFR-β. Further, 11 binds at the colchicine site on tubulin, depolymerizes cellular microtubules and inhibits purified tubulin assembly and overcomes both βIII-tubulin and P-glycoprotein-mediated drug resistance, and initiates mitotic arrest leading to apoptosis. In vivo, its HCl salt, 21, reduced tumor size and vascularity in xenograft and allograft murine models and was superior to docetaxel and sunitinib, without overt toxicity. Thus 21 affords potential combination chemotherapy in a single agent.
Collapse
Affiliation(s)
- Xin Zhang
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States
| | - Sudhir Raghavan
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States
| | - Michael Ihnat
- College of Pharmacy, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Jessica E Thorpe
- College of Pharmacy, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Bryan C Disch
- College of Pharmacy, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Anja Bastian
- College of Pharmacy, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Lora C Bailey-Downs
- College of Pharmacy, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Nicholas F Dybdal-Hargreaves
- Department of Pharmacology, Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Cristina C Rohena
- Department of Pharmacology, Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Institutes of Health, 1050 Boyles Street, Frederick, MD 21702, United States
| | - Susan L Mooberry
- Department of Pharmacology, Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Aleem Gangjee
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States.
| |
Collapse
|
28
|
Bertucci F, Finetti P, Vermeulen P, Van Dam P, Dirix L, Birnbaum D, Viens P, Van Laere S. Genomic profiling of inflammatory breast cancer: a review. Breast 2014; 23:538-45. [PMID: 24998451 DOI: 10.1016/j.breast.2014.06.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 06/02/2014] [Accepted: 06/08/2014] [Indexed: 01/04/2023] Open
Abstract
Inflammatory breast cancer (IBC) is a rare but aggressive form of breast cancer. Despite efforts in the past decade to delineate the molecular biology of IBC by applying high-throughput molecular profiling technologies to clinical samples, IBC remains insufficiently characterized. The reasons for that include limited sizes of the study population, heterogeneity with respect to the composition of the IBC and non-IBC control groups and technological differences across studies. In 2008, the World IBC Consortium was founded to foster collaboration between research groups focusing on IBC. One of the initial projects was to redefine the molecular profile of IBC using an unprecedented number of samples and search for gene signatures associated with survival and response to neo-adjuvant chemotherapy. Here, we provide an overview of all the molecular profiling studies that have been performed on IBC clinical samples to date.
Collapse
Affiliation(s)
- François Bertucci
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes and UMR1068 Inserm, Marseille, France; Département d'Oncologie Médicale, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes and UMR1068 Inserm, Marseille, France; Faculté de Médecine, Université de la Méditerranée, Marseille, France.
| | - Pascal Finetti
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes and UMR1068 Inserm, Marseille, France
| | - Peter Vermeulen
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Wilrijk, Belgium; Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Peter Van Dam
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Wilrijk, Belgium; Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Daniel Birnbaum
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes and UMR1068 Inserm, Marseille, France
| | - Patrice Viens
- Département d'Oncologie Médicale, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes and UMR1068 Inserm, Marseille, France; Faculté de Médecine, Université de la Méditerranée, Marseille, France
| | - Steven Van Laere
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Wilrijk, Belgium; Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
29
|
Mohan A, Ponnusankar S. Newer therapies for the treatment of metastatic breast cancer: a clinical update. Indian J Pharm Sci 2014; 75:251-61. [PMID: 24082340 PMCID: PMC3783742 DOI: 10.4103/0250-474x.117396] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 04/21/2013] [Accepted: 05/01/2013] [Indexed: 12/25/2022] Open
Abstract
Breast cancer is the foremost common malignancy among the female population around the world. Female breast cancer incidence rates have increased since 1980, slowed in 1990, the rate of increase have leveled off since 2001. In spite of the advances in the early detection, treatment, surgery and radiation support, almost 70% of the patients develop metastasis and die of the disease. Around 10% of the patients when diagnosed with breast cancer have metastases. Survival among the breast cancer patients have increased due to the introduction of novel single agent, combination of chemotherapeutic agents and targeted biologic agents, which is breast cancer specific. The staging of tumor-node-metastasis is significant for the prognosis and treatment. Predominantly the combination of chemotherapeutic regimen is given to improve the rate of clinical benefit and the overall survival rate. Novel mono-therapeutic options are being used often in metastatic setting as they will not be able to endure the toxicity of the combination regimen. Usually, endocrine therapy is recommended for hormone-responsive breast cancer due to efficacy and favorable side effect profile but chemotherapy becomes an option when endocrine therapy fails. This review summarizes the newer therapeutic options for early breast cancer and advanced breast cancer that are pretreated heavily on other chemotherapeutic agents. Further it provides monotherapies and other emerging novel combination regime which can be opted for first line or second line setting.
Collapse
Affiliation(s)
- Anjana Mohan
- Department of Pharmacy Practice, JSS College of Pharmacy, The Nilgiris, Ooty-643 001, India
| | | |
Collapse
|
30
|
AZD8931, an equipotent, reversible inhibitor of signaling by epidermal growth factor receptor (EGFR), HER2, and HER3: preclinical activity in HER2 non-amplified inflammatory breast cancer models. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:47. [PMID: 24886365 PMCID: PMC4061513 DOI: 10.1186/1756-9966-33-47] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/13/2014] [Indexed: 12/02/2022]
Abstract
Introduction Epidermal growth factor receptor (EGFR) overexpression has been associated with prognostic and predictive value in inflammatory breast cancer (IBC). Epidermal growth factor receptor 2 (HER2) overexpression is observed at a higher rate in IBC compared with noninflammatory breast cancer. Current clinically available anti-HER2 therapies are effective only in patients with HER2 amplified breast cancer, including IBC. AZD8931 is a novel small-molecule equipotent inhibitor of EGFR, HER2, and HER3 signaling. In this study, we investigated the antitumor activity of AZD8931 alone or in combination with paclitaxel using preclinical models of EGFR-overexpressed and HER2 non-amplified IBC cells. Methods Two IBC cell lines SUM149 and FC-IBC-02 derived from pleural effusion of an IBC patient were used in this study. Cell growth and apoptotic cell death were examined in vitro. For the in vivo tumor growth studies, IBC cells were orthotopically transplanted into the mammary fat pads of immunodeficient mice. AZD8931 was given by daily oral gavage at doses of 25 mg/kg, 5 days/week for 4 weeks. Paclitaxel was subcutaneously injected twice weekly. Results AZD8931 significantly suppressed cell growth of IBC cells and induced apoptosis of human IBC cells in vitro. Significantly, we showed that AZD8931 monotherapy inhibited xenograft growth and the combination of paclitaxel + AZD8931 was demonstrably more effective than paclitaxel or AZD8931 alone treatment at delaying tumor growth in vivo in orthotopic IBC models. Conclusion AZD8931 single agent and in combination with paclitaxel demonstrated signal inhibition and antitumor activity in EGFR-overexpressed and HER2 non-amplified IBC models. These results suggest that AZD8931 may provide a novel therapeutic strategy for the treatment of IBC patients with HER2 non-amplified tumors.
Collapse
|
31
|
Trichostatin A suppresses EGFR expression through induction of microRNA-7 in an HDAC-independent manner in lapatinib-treated cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:168949. [PMID: 24707474 PMCID: PMC3950925 DOI: 10.1155/2014/168949] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/04/2014] [Accepted: 01/06/2014] [Indexed: 12/11/2022]
Abstract
Lapatinib, a dual EGFR/HER2 tyrosine kinase inhibitor, has been shown to improve the survival rate of patients with advanced HER2-positive breast cancers. However, the off-target activity of lapatinib in inducing EGFR expression without tyrosine kinase activity was demonstrated to render HER2-negative breast cancer cells more metastatic, suggesting a limitation to the therapeutic effectiveness of this dual inhibitor in HER2-heterogeneous tumors. Therefore, targeting EGFR expression may be a feasible approach to improve the anticancer efficiency of lapatinib-based therapy. Inhibition of HDAC has been previously reported to epigenetically suppress EGFR protein expression. In this study, however, our data indicated that treatment with HDAC inhibitors trichostatin A (TSA), but not suberoylanilide hydroxamic acid (SAHA) or HDAC siRNA, can attenuate both protein and mRNA expressions of EGFR in lapatinib-treated triple-negative breast cancer cells, suggesting that TSA may suppress EGFR expression independently of HDAC inhibition. Nevertheless, TSA reduced EGFR 3′UTR activity and induced the gene expression of microRNA-7, a known EGFR-targeting microRNA. Furthermore, treatment with microRNA-7 inhibitor attenuated TSA-mediated EGFR suppression. These results suggest that TSA induced microRNA-7 expression to downregulate EGFR expression in an HDAC-independent manner.
Collapse
|
32
|
Karki R, Seagle BLL, Nieves-Neira W, Shahabi S. Taxanes in combination with biologic agents for ovarian and breast cancers. Anticancer Drugs 2013; 25:536-54. [PMID: 24300916 DOI: 10.1097/cad.0000000000000056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Taxane-based cytotoxic therapy is commonly prescribed for breast and ovarian cancers. Although these cancers are often sensitive to such therapy, clinical benefit and overall survival are limited owing to the development of chemoresistance and recurrence. Biologic agents that specifically target proteins of growth factor signaling pathways, which are hyperactivated in cancers, offer attractive targets for cancer therapeutics and may work synergistically with standard taxane-based chemotherapy to improve patient outcomes. We review clinical trials of biologic agents--angiogenic, tyrosine kinase, and antibody inhibitors--in combination with taxane-based therapy for ovarian and breast cancers. Many clinical trials have shown promising results. However, some biologic agents still need larger trials to assess safety and efficacy. As research into the heterogeneity and complexity of ovarian and breast cancers improves our understanding of the molecular pathways involved, there is no question that targeted therapies with biologic agents will expand the future array of available cancer therapeutics.
Collapse
Affiliation(s)
- Roshan Karki
- aReproductive Tumor Biology Research, Department of Obstetrics and Gynecology, Danbury Hospital, Danbury, Connecticut bDivision of Gynecologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | | | | | | |
Collapse
|
33
|
Azim HA, Agbor-Tarh D, Bradbury I, Dinh P, Baselga J, Di Cosimo S, Greger JG, Smith I, Jackisch C, Kim SB, Aktas B, Huang CS, Vuylsteke P, Hsieh RK, Dreosti L, Eidtmann H, Piccart M, de Azambuja E. Pattern of rash, diarrhea, and hepatic toxicities secondary to lapatinib and their association with age and response to neoadjuvant therapy: analysis from the NeoALTTO trial. J Clin Oncol 2013; 31:4504-11. [PMID: 24248687 DOI: 10.1200/jco.2013.50.9448] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
PURPOSE We investigated the pattern of rash, diarrhea, and hepatic adverse events (AEs) secondary to lapatinib and their association with age and pathologic complete response (pCR) in the Neoadjuvant Lapatinib and/or Trastuzumab Treatment Optimisation (NeoALLTO) phase III trial. PATIENTS AND METHODS Patients with HER2-positive early breast cancer were randomly assigned to receive lapatinib (Arm A), trastuzumab (Arm B), or their combination (Arm C) for 6 weeks followed by the addition of paclitaxel for 12 weeks before surgery. We investigated the frequency and time to developing each AE according to age (≤ 50 v > 50 years) and their association with pCR in a logistic regression model adjusted for age, hormone receptors, tumor size, nodal status, planned breast surgery, completion of lapatinib administration, and treatment arm. RESULTS Only patients randomly assigned to arms A and C were eligible (n = 306). Younger patients (≤ 50 years) experienced significantly more rash compared with older patients (74.4% v 47.9%; P < .0001). Diarrhea and hepatic AEs were observed in 78.8% and 41.2% of patients, respectively, with no differences in rate or severity or time of onset according to age. Early rash (ie, before starting paclitaxel) was independently associated with a higher chance of pCR, mainly in patients older than 50 years (odds ratio [OR] = 3.76; 95% CI, 1.69 to 8.34) but not in those ≤ 50 years (OR = 0.92; 95% CI, 0.45 to 1.88; P for interaction = .01). No significant association was observed between pCR and diarrhea or hepatic AEs. CONCLUSION Our results indicate that the frequency and clinical relevance of lapatinib-related rash is largely dependent on patient age.
Collapse
Affiliation(s)
- Hatem A Azim
- Jose Baselga, Memorial Sloan-Kettering Cancer Center, NY; James G. Greger Jr, GlaxoSmithKline, Collegeville, PA; Hatem A. Azim Jr, Martine Piccart, Evandro de Azambuja, BrEAST Data Centre, Institut Jules Bordet, Université Libre de Bruxelles; Phuong Dinh, Breast International Group, Brussels; Peter Vuylsteke, Sint-Elisabeth Hospital, Namur, Belgium; Dominique Agbor-tarh, Ian Bradbury, Frontier Science, Kincraig, Kingussie, Scotland; Ian Smith, Royal Marsden Hospital, and Institute of Cancer Research, London, United Kingdom; Serena Di Cosimo, IRCCS Fondazione Instituto Nazionale dei Tumori, Milan, Italy; Serena Di Cosimo, SOLTI Breast Cancer Research Group, Barcelona, Spain; Christian Jackisch, Kilinikum Offenbach, Offenbach; Bahriye Aktas, Kliniken Essen-Mitte, Evang, Essen; Holger Eidtmann, University Hospital Kiel, Kiel, Germany; Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea; Chiun-Sheng Huang, National Taiwan University Hospital; Ruey Kuen Hsieh, Mackey Memorial Hospital, Taipei, Taiwan; and Lydia Dreosti, University of Pretoria, Pretoria, South Africa
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Chen YJ, Yeh MH, Yu MC, Wei YL, Chen WS, Chen JY, Shih CY, Tu CY, Chen CH, Hsia TC, Chien PH, Liu SH, Yu YL, Huang WC. Lapatinib-induced NF-kappaB activation sensitizes triple-negative breast cancer cells to proteasome inhibitors. Breast Cancer Res 2013; 15:R108. [PMID: 24216290 PMCID: PMC3979035 DOI: 10.1186/bcr3575] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 10/31/2013] [Indexed: 12/27/2022] Open
Abstract
Introduction Triple-negative breast cancer (TNBC), a subtype of breast cancer with negative expressions of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2), is frequently diagnosed in younger women and has poor prognosis for disease-free and overall survival. Due to the lack of known oncogenic drivers for TNBC proliferation, clinical benefit from currently available targeted therapies is limited, and new therapeutic strategies are urgently needed. Methods Triple-negative breast cancer cell lines were treated with proteasome inhibitors in combination with lapatinib (a dual epidermal growth factor receptor (EGFR)/HER2 tyrosine kinase inhibitor). Their in vitro and in vivo viability was examined by MTT assay, clonogenic analysis, and orthotopic xenograft mice model. Luciferase reporter gene, immunoblot, and RT-qPCR, immunoprecipitation assays were used to investigate the molecular mechanisms of action. Results Our data showed that nuclear factor (NF)-κB activation was elicited by lapatinib, independent of EGFR/HER2 inhibition, in TNBCs. Lapatinib-induced constitutive activation of NF-κB involved Src family kinase (SFK)-dependent p65 and IκBα phosphorylations, and rendered these cells more vulnerable to NF-κB inhibition by p65 small hairpin RNA. Lapatinib but not other EGFR inhibitors synergized the anti-tumor activity of proteasome inhibitors both in vitro and in vivo. Our results suggest that treatment of TNBCs with lapatinib may enhance their oncogene addiction to NF-κB, and thus augment the anti-tumor activity of proteasome inhibitors. Conclusions These findings suggest that combination therapy of a proteasome inhibitor with lapatinib may benefit TNBC patients.
Collapse
|
35
|
Hsia TC, Tu CY, Chen YJ, Wei YL, Yu MC, Hsu SC, Tsai SL, Chen WS, Yeh MH, Yen CJ, Yu YL, Huang TC, Huang CY, Hung MC, Huang WC. Lapatinib-mediated cyclooxygenase-2 expression via epidermal growth factor receptor/HuR interaction enhances the aggressiveness of triple-negative breast cancer cells. Mol Pharmacol 2013; 83:857-69. [PMID: 23355539 DOI: 10.1124/mol.112.082743] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Lapatinib, a dual epidermal growth factor receptor (EGFR)/human epidermal growth factor receptor 2 (HER2) kinase inhibitor, showed clinical benefits in advanced HER2-positive breast cancer patients. Because some triple-negative breast cancers (TNBCs) frequently overexpress EGFR, the antitumor activity of lapatinib in such diseases was also tested. However, the results showed a worse event-free survival rate. It remains unknown whether and how lapatinib elicits the aggressiveness of such cancer cells. In this study, our results demonstrated that lapatinib facilitated axillary and lung metastases of triple-negative MDA-MB-231 breast cancer cells without affecting their viability, leading to worse survival in orthotopic xenograft mice. The lapatinib-increased motility was attributed by the elevation of EGFR through the downregulation of microRNA-7 and by the subsequent overexpression of cyclooxygenase-2 (COX-2). Strikingly, independent of its kinase activity, the elevated EGFR at least partly stabilized COX-2 expression by enhancing the binding of HuR to COX-2 mRNA. Our results suggest that lapatinib may increase the migration and invasion of MDA-MB-231 cells by upregulating EGFR and COX-2 through the downregulation of microRNA-7, providing a potential explanation for the worse clinical outcome of TNBC patients who receive lapatinib-based treatment. These findings also shed new light on the molecular mechanism of COX-2 mRNA stabilization by EGFR in a kinase-independent manner.
Collapse
Affiliation(s)
- Te-Chun Hsia
- Department of Internal Medicine, China Medical University and Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Dent S, Oyan B, Honig A, Mano M, Howell S. HER2-targeted therapy in breast cancer: a systematic review of neoadjuvant trials. Cancer Treat Rev 2013; 39:622-31. [PMID: 23434074 DOI: 10.1016/j.ctrv.2013.01.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 01/07/2013] [Accepted: 01/10/2013] [Indexed: 12/18/2022]
Abstract
Targeting human epidermal growth factor receptor 2 (HER2) during or in sequence with chemotherapy improves overall survival in metastatic and early HER2-overexpressing breast cancer. In this paper we systematically review neoadjuvant clinical trial data in HER2-positive breast cancer and discuss key unanswered clinical questions. All trials of HER2-targeted neoadjuvant therapy were identified through non-date-limited searches of PubMED® and Biosis® and congress abstract book searches from 2000-2011. Eligible trials were prospective, had at least 10 patients and a clear definition of pathological complete response (pCR) rate. A total of 50 trials fulfilled the eligibility criteria; 41 single-arm phase II studies were identified, 37 with trastuzumab and 4 with lapatinib, with significant variability in baseline tumour characteristics and pCR rates (range 12-66.7%). Of 9 randomised phase II/III trials, 4 assessed the addition of trastuzumab to chemotherapy and a further 5 randomised trials assessed different HER2-targeting approaches. Four of these studies assessed dual HER2-targeting approaches, which universally increased pCR at the expense of increased non-cardiac toxicity when lapatinib, but not pertuzumab, was added to trastuzumab. Significant advances have been made in HER2 targeting, resulting in a marked increase in the number of breast cancer patients experiencing tumour pCR. Mature data from randomised neoadjuvant and adjuvant studies are awaited for survival outcomes with combination targeted approaches. Unanswered questions centre on the individualisation of therapy and include; which, if any, chemotherapy backbone should be used, and which patients need dual HER2 blockade?
Collapse
Affiliation(s)
- Susan Dent
- The Ottawa Hospital Cancer Centre, Division of Medical Oncology, Department of Medicine, The University of Ottawa, 501 Smyth Road, Box 912, Ottawa, Ontario, Canada.
| | | | | | | | | |
Collapse
|
37
|
A randomized phase II study of lapatinib + pazopanib versus lapatinib in patients with HER2+ inflammatory breast cancer. Breast Cancer Res Treat 2012; 137:471-82. [PMID: 23239151 PMCID: PMC3539065 DOI: 10.1007/s10549-012-2369-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 11/30/2012] [Indexed: 01/04/2023]
Abstract
This multi-center Phase II study evaluated lapatinib, pazopanib, and the combination in patients with relapsed HER2+ inflammatory breast cancer. In Cohort 1, 76 patients were randomized 1:1 to receive lapatinib 1,500 mg + placebo or lapatinib 1,500 mg + pazopanib 800 mg (double-blind) once daily until disease progression, unacceptable toxicity, or death. Due to high-grade diarrhea observed with this dose combination in another study (VEG20007), Cohort 1 was closed. The protocol was amended such that an additional 88 patients (Cohort 2) were randomized in a 5:5:2 ratio to receive daily monotherapy lapatinib 1,500 mg, lapatinib 1,000 mg + pazopanib 400 mg, or monotherapy pazopanib 800 mg, respectively. The primary endpoint was overall response rate (ORR). Secondary endpoints included duration of response, progression-free survival (PFS), overall survival, and safety. In Cohort 1, ORR for the lapatinib (n = 38) and combination (n = 38) arms was 29 and 45 %, respectively; median PFS was 16.1 and 14.3 weeks, respectively. Grade ≥3 adverse events (AEs) were more frequent in the combination arm (71 %) than in the lapatinib arm (24 %). Dose reductions and interruptions due to AEs were also more frequent in the combination arm (45 and 53 %, respectively) than in the lapatinib monotherapy arm (0 and 11 %, respectively). In Cohort 2, ORR for patients treated with lapatinib (n = 36), lapatinib + pazopanib (n = 38), and pazopanib (n = 13) was 47, 58, and 31 %, respectively; median PFS was 16.0, 16.0, and 11.4 weeks, respectively. In the lapatinib, combination, and pazopanib therapy arms, grade ≥3 AEs were reported for 17, 50, and 46 % of patients, respectively, and the incidence of discontinuations due to AEs was 0, 24, and 23 %, respectively. The lapatinib-pazopanib combination was associated with a numerically higher ORR but no increase in PFS compared to lapatinib alone. The combination also had increased toxicity resulting in more dose reductions, modifications, and treatment delays. Activity with single-agent lapatinib was confirmed in this population.
Collapse
|
38
|
New Therapeutic Targets in Inflammatory Breast Cancer. CURRENT BREAST CANCER REPORTS 2012. [DOI: 10.1007/s12609-012-0087-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
39
|
Singla S, Pippin JA, Drebin JA. Dual ErbB1 and ErbB2 receptor tyrosine kinase inhibition exerts synergistic effect with conventional chemotherapy in pancreatic cancer. Oncol Rep 2012; 28:2211-6. [PMID: 23007710 DOI: 10.3892/or.2012.2053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 07/09/2012] [Indexed: 11/05/2022] Open
Abstract
Patient survival in pancreatic cancer remains poor with gemcitabine (GEM)-based regimens. The target specific molecular agent lapatinib, a dual ErbB1 and ErbB2 receptor tyrosine kinase inhibitor, has shown significant activity against ErbB1 and ErbB2-expressing tumors. Since pancreatic tumors frequently overexpress these proteins, we investigated its effects, both alone and in conjunction with 5-FU or GEM. The pancreatic cancer cell lines PANC-1 and AsPC were treated with varying doses of lapatinib in vitro. The effects on ErbB1/ErbB2 protein phosphorylation and on the cell survival protein survivin were determined by western blotting. Cytotoxicity was determined by MTT assay and apoptosis was measured using the caspase-3 colorimetric assay. Similar dose-response lapatinib experiments were conducted with varying concentrations of 5-FU or GEM and isobolograms were constructed to evaluate therapeutic synergy. Lapatinib inhibited protein phosphorylation in the range of 4-16 µM, a clinically achievable concentration. The lapatinib-treated cells showed a dose-dependent inhibition of cell proliferation and induction of apoptosis at the same concentrations that blocked ErbB1/ErbB2 phosphorylation. The addition of 5-FU or GEM to these cells resulted in synergistic effects. The lapatinib-treated cells also demonstrated downregulation of survivin. Simultaneous dual ErbB1 and ErbB2 receptor tyrosine kinase inhibition with lapatinib results in significant reduction of pancreatic cancer cell growth and proliferation. These effects occur at clinically achievable concentrations and are synergistic with the effects of 5-FU or GEM. These findings support the potential role of lapatinib in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Smit Singla
- Department of Surgery, Temple University Hospital, Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|
40
|
Aissi S, Ben Mrad M, Zarraa S, Bounedjar A, Laabidi S, Boussen H. [Targeted therapies: towards a new toxicology?]. PATHOLOGIE-BIOLOGIE 2012; 60:234-238. [PMID: 22743098 DOI: 10.1016/j.patbio.2012.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 05/14/2012] [Indexed: 06/01/2023]
Abstract
Targeted therapies (TT) represent since 10 years, an interesting progress in oncology for many cancers in adjuvant, neoadjuvant or palliative situation. The development of this new class of drugs, with an original mechanism of action, their easy administration, mainly per os, and a particular toxicity profile different from "classical" chemotherapy (CT) leads them entering in the therapeutic arsenal of breast, digestive tract, lung and hematologic cancers, in association with CT. Medical oncologists took rapidly the train of TT, managing a new and original skin, digestive, cardiovascular, endocrine and pulmonary toxicity profile, that remains relatively less important than "classical" CT.
Collapse
Affiliation(s)
- S Aissi
- Service de carcinologie médicale, hôpital Abderrahman-Mami, rue Abderrahmen-Mami, 2080 Ariana-Tunis, Tunisie
| | | | | | | | | | | |
Collapse
|
41
|
Scotti V, Desideri I, Meattini I, Di Cataldo V, Cecchini S, Petrucci A, Franzese C, Greto D, Livi L, Bonomo P, Biti G. Management of inflammatory breast cancer: focus on radiotherapy with an evidence-based approach. Cancer Treat Rev 2012; 39:119-24. [PMID: 22564719 DOI: 10.1016/j.ctrv.2012.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 04/03/2012] [Indexed: 11/29/2022]
Abstract
Inflammatory breast cancer represents a rare and extremely aggressive subtype of breast cancer. Due to its rarity, prospective studies are a difficult goal to obtain in this field. Nowadays a multimodal approach seems to be the standard approach. Role and timing of surgery, radiotherapy and chemotherapy are still debated issues. In this scenario interest is rising in molecular and target therapies. We performed a review analyzing the management of this unfavorable disease focusing on the role of radiotherapy, with particular emphasis on levels of evidence.
Collapse
Affiliation(s)
- Vieri Scotti
- Department of Radiation-Oncology, University of Florence, Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hurvitz SA, Kakkar R. Role of lapatinib alone or in combination in the treatment of HER2-positive breast cancer. BREAST CANCER-TARGETS AND THERAPY 2012; 4:35-51. [PMID: 24367193 DOI: 10.2147/bctt.s29996] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE This review aims to present the preclinical and clinical data regarding efficacy and safety of lapatinib alone and in combination with other agents in the treatment of human epidermal growth factor receptor-2 (HER2)-overexpressing breast cancer. BACKGROUND HER2-positive (HER2+) breast cancer remains a treatment challenge. It is more aggressive than other breast cancers and it is associated with a poor outcome. Targeted therapy for HER2+ breast cancer has significantly changed the clinical course of the disease. Despite advances in therapy, there remains an unmet need in the treatment of HER2+ breast cancer. Lapatinib is a novel, orally bioavailable epidermal growth factor receptor/HER2+ targeted agent. Many trials have investigated the efficacy and safety of lapatinib alone and in conjunction with other agents in the treatment of HER2+ breast cancer. METHODS AND RESULTS Preclinical and clinical trials of lapatinib have shown that it is effective in the treatment on HER2+ breast cancer. More important, studies show that it is effective in the setting of trastuzumab resistance and in the treatment of central nervous system metastases, both of which are current treatment challenges. Furthermore, lapatinib is effective in conjunction with trastuzumab in the treatment of early breast cancer. Data regarding the safety of lapatinib show that it is generally well tolerated; however, multiple studies have shown significant (grade 3 and 4) diarrhea and rash associated with lapatinib, thereby limiting its use. Carditoxicity has not been a significant adverse event associated with the use of lapatinib. CONCLUSION Lapatinib is effective alone and in conjunction with other agents in the treatment of HER2+ breast cancer. However, its use is limited by significant diarrhea and rash.
Collapse
Affiliation(s)
- Sara A Hurvitz
- Department of Medicine, University of California, Los Angeles, CA, USA
| | - Reva Kakkar
- Department of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
43
|
Abstract
Preventing breast cancer is possible with selective estrogen receptor (ER) modulators and aromatase inhibitors, which reduce the risk of invasive disease by up to 65% (up to 73% for ER-positive and no effect for ER-negative cancer) and the risk of preinvasive disease [ductal carcinoma in situ (DCIS)] by up to 50%. Clearly, approaches for preventing ER-negative, and increased prevention of ER-positive breast cancers would benefit public health. A growing body of work (including recent preclinical and clinical data) support targeting the HER family [epidermal growth factor receptor (EGFR), or human epidermal growth factor receptor (HER) 1 or ErbB1) and HER2, HER3, and HER4] for preventing ER-negative and possibly ER-positive breast cancer. Preclinical studies of HER family-targeting drugs in mammary neoplasia show suppression of (i) ER-negative tumors in HER2-overexpressing mouse strains, (ii) ER-negative tumors in mutant Brca1/p53(+/-) mice, and (iii) ER-positive tumors in the methylnitrosourea (MNU) rat model; tumors arising in both the MNU and mutant Brca1/p53(+/-) models lack HER2 overexpression. Clinical trials include a recent placebo-controlled phase IIb presurgical trial of the dual EGFR HER2 inhibitor lapatinib that suppressed growth of breast premalignancy [including atypical ductal hyperplasia (ADH) and DCIS] and invasive cancer in patients with early-stage, HER2-overexpressing or -amplified breast cancer. These results suggest that lapatinib can clinically suppress the progression of ADH and DCIS to invasive breast cancer, an effect previously observed in a mouse model of HER2-overexpressing, ER-negative mammary cancer. The preclinical and clinical signals provide a compelling rationale for testing HER-targeting drugs for breast cancer prevention in women at moderate-to-high risk, leading perhaps to combinations that prevent ER-negative and ER-positive breast cancer.
Collapse
Affiliation(s)
- Louise R Howe
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, USA
| | | |
Collapse
|
44
|
Chaher N, Arias-Pulido H, Terki N, Qualls C, Bouzid K, Verschraegen C, Wallace A, Royce M. Molecular and epidemiological characteristics of inflammatory breast cancer in Algerian patients. Breast Cancer Res Treat 2012; 131:437-44. [PMID: 21360074 PMCID: PMC3564504 DOI: 10.1007/s10549-011-1422-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 02/21/2011] [Indexed: 01/04/2023]
Abstract
Inflammatory breast cancer (IBC) shows a high incidence in Tunisia and Egypt but epidemiological and molecular characteristics have not been described in Algeria. We compared 117 IBC and 59 non-IBC locally advanced breast cancers (LABC), for estrogen and progesterone receptors, HER2, and EGFR protein expression by immunohistochemistry, and HER2 gene amplification by chromogenic in situ hybridization. Demographic, clinico-pathological, and molecular variables were compared with chi-square and Fisher's exact tests to test for significance (P < 0.05, two-tailed). Overall survival (OS) and disease-free survival (DFS) were plotted using Kaplan-Meier curves and compared using the log-rank test. Tumor emboli were detected in 77% of IBC. Palpable masses were found in all LABC but only in 32% of IBC (P < 0.001). Recurrences were higher in LABC than in IBC (48 vs. 35%; P = 0.14) but OS was worse in IBC (68 vs. 71%; P = 0.06). There were no significant differences between IBC and LABC by demographics or by clinico-pathological parameters. The majority of IBC and LABC tumors were luminal A (62 and 64%), followed by basal (~18%, each), triple negative (~18%, each), and HER2+ (~10%, each) subtypes. In multivariate analyses, grade was associated with worse OS (P = 0.04), and DFS (P < 0.001) in IBC; chemo- and radio-therapy were associated with improved OS and DFS, respectively (P < 0.05 for each) in LABC. In conclusion, IBC in Algeria shows similar characteristics to IBC described for Egypt and Tunisia with subtle molecular differences. Current therapeutic treatments were not very effective in this population and new approaches are much needed.
Collapse
Affiliation(s)
- Nabila Chaher
- Department of Pathology, Centre Pierre et Marie Curie, 1, Avenue Battendier, Place May 1, Algiers, Algeria
| | - Hugo Arias-Pulido
- Clinical Research and Translational Therapeutics. The University of New Mexico Cancer Center, Albuquerque, NM, USA
| | - Nadija Terki
- Department of Pathology, Centre Pierre et Marie Curie, 1, Avenue Battendier, Place May 1, Algiers, Algeria
| | - Clifford Qualls
- Department of Mathematics and Statistics. The University of New Mexico Cancer Center, Albuquerque, NM, USA
| | - Kamel Bouzid
- Department of Medical Oncology, Centre Pierre et Marie Curie, 1, Avenue Battendier, Place May 1, Algiers, Algeria
| | - Claire Verschraegen
- Clinical Research and Translational Therapeutics. The University of New Mexico Cancer Center, Albuquerque, NM, USA
| | - AnneMarie Wallace
- Department of Internal Medicine. The University of New Mexico Cancer Center, Albuquerque, NM, USA
| | - Melanie Royce
- Department of Internal Medicine. The University of New Mexico Cancer Center, Albuquerque, NM, USA
| |
Collapse
|
45
|
The role of lapatinib in the preoperative therapy of breast cancer. Curr Oncol Rep 2011; 14:44-7. [PMID: 22037924 DOI: 10.1007/s11912-011-0206-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Preoperative or neoadjuvant chemotherapy is a well-established modality in the treatment of nonmetastatic breast cancer. Patients initially considered inoperable may be able to achieve operable status after preoperative chemotherapy and patients initially considered not to be candidates for breast conservation may convert to breast conservation status. Human epidermal growth factor receptor 2 (Her2)-positive tumors have been shown to have a more aggressive course including early local relapse and metastasis when compared to Her2-negative breast cancers, but the optimal use of Her2-targeted agents is constantly evolving as new agents become available. Preoperative studies allow us to quickly assess the activity of new agents and combinations for particular biological subsets of breast cancer.
Collapse
|
46
|
Semiglazov V, Eiermann W, Zambetti M, Manikhas A, Bozhok A, Lluch A, Tjulandin S, Sabadell MD, Caballero A, Valagussa P, Baselga J, Gianni L. Surgery following neoadjuvant therapy in patients with HER2-positive locally advanced or inflammatory breast cancer participating in the NeOAdjuvant Herceptin (NOAH) study. Eur J Surg Oncol 2011; 37:856-63. [PMID: 21843921 DOI: 10.1016/j.ejso.2011.07.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 06/29/2011] [Accepted: 07/25/2011] [Indexed: 11/18/2022] Open
Abstract
AIM To describe surgical outcomes in patients with HER2-positive locally advanced (LABC) or inflammatory breast cancer (IBC) participating in the NeOAdjuvant Herceptin (NOAH) study (ISRCTN86043495). PATIENTS AND METHODS A total of 235 patients with HER2-positive disease were randomized to neoadjuvant trastuzumab plus chemotherapy (doxorubicin plus paclitaxel, followed by paclitaxel, followed by cyclophosphamide, methotrexate and fluorouracil) or neoadjuvant chemotherapy alone. Of these patients, 228 received their allocated treatment (115 received trastuzumab plus chemotherapy and 113 received chemotherapy alone) and were potentially eligible for surgery. Mastectomy was required for all patients with IBC and was recommended for all patients with LABC. However, breast-conserving therapy could be considered for patients with peripheral neoplasms measuring ≤ 4 cm in diameter at diagnosis, with a favorable ratio of tumor to breast volume, or at the patient's request if there had been a good response to treatment. RESULTS As previously reported, the addition of trastuzumab to neoadjuvant chemotherapy improved the overall, complete and pathological complete response to therapy and significantly improved event-free survival (the primary endpoint of the study). Trastuzumab also enabled more patients to have breast conserving surgery (BCS) (23% versus 13% respectively) without an apparent detrimental effect on local disease control (no patient treated with trastuzumab plus chemotherapy had experienced a local recurrence after BCS at the time of analysis). CONCLUSIONS Although this was not an aim of the trial, neoadjuvant trastuzumab given concurrently with chemotherapy enabled 23% of patients with HER2-positive LABC/IBC to avoid mastectomy (including a small number of patients with IBC).
Collapse
Affiliation(s)
- V Semiglazov
- NN Petrov Research Institute of Oncology, St. Petersburg Oncology, 68, Leningradskaya Str. Pesochny-2, St. Petersburg, Russian Federation, Russia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Hassan SE, Bekarev M, Kim MY, Lin J, Piperdi S, Gorlick R, Geller DS. Cell surface receptor expression patterns in osteosarcoma. Cancer 2011; 118:740-9. [PMID: 21751203 DOI: 10.1002/cncr.26339] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 05/16/2011] [Accepted: 05/18/2011] [Indexed: 01/17/2023]
Abstract
BACKGROUND Although the presence of numerous cell signaling receptors in osteosarcoma is known, their simultaneous characterization has not been performed to date. The current study sought to characterize and quantify the expression of cell surface receptors across a variety of osteosarcoma cell lines. METHODS Standard (n = 4) and patient-derived (n = 10) osteosarcoma cell lines were cultured and labeled with antibodies to epidermal growth factor receptor, human epidermal growth factor receptor (HER)-2, HER-3, HER-4, insulin-like growth factor 1 receptor (IGF-1R), IGF-2R, insulin receptor (IR), vascular endothelial growth factor receptor (VEGFR)-1, VEGFR-2, VEGFR-3, c-Met, fibroblast growth factor receptor (FGFR)-2, FGFR-3, and platelet-derived growth factor receptor (PDGFR)-β. Cell surface examination was performed using flow cytometry, and the geometric fluorescent mean for each receptor was calculated and compared against a positive control. RESULTS Significant overexpression of IGF-2R was shown in all cell lines, with an average geometric mean above the upper expression quartile. A variable expression pattern was seen for c-Met, PDGFR-β, IR, IGFR-1, HER-2, and VEGFR-3 with expression values for the remaining receptors mainly in the lower quartile. An apparent association between the expression of IGF-1R and HER-2 and between the expression of PDGFR-β and IR was demonstrated. CONCLUSION IGF-2R was consistently overexpressed on the cell surface across all tested osteosarcoma cell lines. Substantial, although variable, expression of c-Met, HER-2, IGF-1R, VEGFR-3, IR, and PDGFR-β was demonstrated as well, suggesting that these receptors may contribute to osteosarcoma aggressiveness and biological heterogeneity and may serve as potential targets within a subset of tumors. Associated receptor expression may provide new insight into common regulatory factors or pathways. Targeting either common factors or targeting multiple specific receptors may have therapeutic relevance.
Collapse
Affiliation(s)
- Sheref E Hassan
- Department of Orthopaedic Surgery, Montefiore Medical Center and The Children's Hospital at Montefiore, Bronx, New York 10467, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Bouchalova K, Cizkova M, Cwiertka K, Trojanec R, Friedecky D, Hajduch M. Lapatinib in breast cancer - the predictive significance of HER1 (EGFR), HER2, PTEN and PIK3CA genes and lapatinib plasma level assessment. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2011; 154:281-8. [PMID: 21293538 DOI: 10.5507/bp.2010.043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Breast cancer treatment trends are currently based on tailored therapies using tumor and patient biomarkers. Lapatinib is the first dual inhibitor of HER1 (EGFR, ErbB1) and HER2 (ErbB2, Neu) tyrosine kinases to be used in clinical practice. However, only HER2 is currently used for therapy indications and new predictors for the treatment with lapatinib are sought. METHODS AND RESULTS This minireview focuses on lapatinib and its role in breast cancer treatment. Preclinical and clinical studies as well as pharmacological characteristics are briefly reviewed while the focus is on efficacy assessment including predictive factors for therapy outcome. CONCLUSION Lapatinib (Tykerb/Tyverb) was Food and Drug Administration (FDA) approved in 2007 for use in combination with capecitabine for the treatment of HER2-positive advanced or metastatic breast cancer in patients who had received previous treatment (including anthracycline, taxane and trastuzumab containing regimens) and in 2010 for use in combination with letrozole for postmenopausal women with hormonal receptor positive and HER2- positive metastatic breast cancer. In contrast to trastuzumab (Herceptin), lapatinib is orally administered and it targets both HER2 and HER1 receptors. As a synthetic and oral tyrosine kinase inhibitor (TKI), it is convenient, cheaper and easier to produce than monoclonal antibodies. The recommended dosage is not dependent on body weight either. Lapatinib plasma level measurement could be an approach to tailored therapy for further optimizing the dose and prolonging this efficient therapy. New lapatinib response predictors are being evaluated. At this time, only HER2 amplification/overexpression is used to choose lapatinib therapy candidates. Further studies on concurrent HER1 fluorescent in situ hybridization (FISH)/immunohistochemistry (IHC) assessment and/or microarray analyses may produce new data on the predictive role of the HER1 (EGFR) gene/protein. PTEN loss and PIK3CA gene mutations are other markers that may predict lapatinib poor response.
Collapse
Affiliation(s)
- Katerina Bouchalova
- Laboratory of Experimental Medicine, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Czech Republic.
| | | | | | | | | | | |
Collapse
|
49
|
Pilot neoadjuvant trial in HER2 positive breast cancer with combination of nab-paclitaxel and lapatinib. Breast Cancer Res Treat 2011; 132:833-42. [DOI: 10.1007/s10549-011-1411-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 02/13/2011] [Indexed: 10/18/2022]
|
50
|
Geuna E, Milani A, Redana S, Rossi V, Valabrega G, Aglietta M, Montemurro F. Hitting multiple targets in HER2-positive breast cancer: proof of principle or therapeutic opportunity? Expert Opin Pharmacother 2011; 12:549-65. [DOI: 10.1517/14656566.2011.525218] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|