1
|
Gao L, Zhang Y, Feng M, Shen M, Yang L, Wei B, Zhou Y, Zhang Z. HER3: Updates and current biology function, targeted therapy and pathologic detecting methods. Life Sci 2024; 357:123087. [PMID: 39366553 DOI: 10.1016/j.lfs.2024.123087] [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: 07/20/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/06/2024]
Abstract
Being a member of the EGFR tyrosine kinase family, HER3 has been shown to be overexpressed in a number of cancers, including breast cancer (BC). The kinase activity of HER3 is extremely low, and it forms heterodimers with partners, HER2 in particular, that promote biological processes like cell migration, survival, and proliferation by activating downstream carcinogenic signaling pathways. The overexpression of HER3 is also directly linked to tumor invasion, metastasis, and a poor prognosis. Despite the relatively low expression of HER3 compared to EGFR and HER2, a lot of targeted drugs are making their way into clinical trials and seem to have a bright further. This review aims to summarize the relationship between HER3 overexpression, mutations, and carcinogenicity and drug resistance, starting from the unique structure and kinase activity of HER3. Simultaneously, numerous approaches to HER3 targeted therapy are enumerated, and the clinical detection methods for HER3 that are commonly employed in pathology are sorted and contrasted to offer physicians a range of options. We think that a better knowledge of the mechanisms underlying HER3 in tumors and the advancement of targeted HER3 therapy will contribute to an improved prognosis for cancer patients and an increase in the efficacy of anticancer therapies.
Collapse
Affiliation(s)
- Leyi Gao
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Yu Zhang
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Mengna Feng
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Mengjia Shen
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Libo Yang
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Bing Wei
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Yongjie Zhou
- Laboratory of Liver Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Zhang Zhang
- Department of Pathology, West China Hospital, Sichuan University, No.37, Guo Xue Xiang, Chengdu 610041, Sichuan, China.
| |
Collapse
|
2
|
Okwor V, Okwor CJ, Ukwuoma M, Nweke M. Effectiveness of combined targeted and hormonal therapies for post-menopausal women with hormone receptor-positive and HER2-negative advanced breast cancer: A systematic review and meta-analysis of RCTs. J Oncol Pharm Pract 2024:10781552241279019. [PMID: 39295518 DOI: 10.1177/10781552241279019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
OBJECTIVE we aim to synthesize available evidence on the effectiveness of hormonal plus targeted therapies for post-menopausal women with hormone receptor-positive and HER2-negative advanced breast cancer. DATA SOURCES AND METHODS We searched the following databases: Medline, PubMed, Cochrane Library, CINAHL, Web of Science, Scopus, and African Journal. Only studies that investigated the effectiveness of hormonal therapy combined with targeted therapy for HR+/HER2- advanced breast cancer treatment were included. The outcomes were progression-free survival (PFS), overall survival (OS) and objective response rate (ORR). A random-effect meta-analysis model was employed. Statistical analysis was performed using Comprehensive Meta-analysis version 3. RESULTS 24 studies were included in the meta-analysis with an overall sample size of 7635. Median PFS, OS and ORR were found to be significantly increased in the combination group compared to hormonal monotherapy [SMD = 6.072 (95% CI = 3.785-8.360), p < 0.001], [SMD = 1.614 (95% CI = 0.139-3.089), p = 0.032] and [OR = 1.584 (CI 1.134-2.213), p = 0.007] respectively. Subgroup analysis showed a significant difference in PFS and ORR between patients who received "hormonal therapy + CDK4/6 inhibitors" vs hormonal therapy only [SMD = 6.015 (CI 3.069-8.960), p < 0.001], (OR = 1.828 (CI 1.030-3.243), p = 0.039] respectively. CONCLUSION Compared with hormonal monotherapy, targeted plus hormonal therapy significantly improves PFS, OS and ORR in postmenopausal women with HR+/HER2- advanced breast cancer.
Collapse
Affiliation(s)
- Vitalis Okwor
- Department of Radiation and Clinical Oncology, University of Nigeria Teaching Hospital, Enugu, Nigeria
| | - Chika Juliet Okwor
- Department of Chemical Pathology, College of Medicine, University of Nigeria Ituku-Ozalla Campus, Enugu, Nigeria
| | - Maryjane Ukwuoma
- Department of Physiotherapy, University of Nigeria Teaching Hospital, Enugu, Enugu
| | - Martins Nweke
- Department of Physiotherapy, Evangel University Akaeze, Enugu, Nigeria
| |
Collapse
|
3
|
Qi Y, Deng SM, Wang KS. Receptor tyrosine kinases in breast cancer treatment: unraveling the potential. Am J Cancer Res 2024; 14:4172-4196. [PMID: 39417188 PMCID: PMC11477839 DOI: 10.62347/kivs3169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Breast cancer is a multifactorial disease driven by acquired genetic and epigenetic changes that lead to aberrant regulation of cellular signaling pathways. Receptor tyrosine kinases (RTKs), a class of critical receptors, are involved in the initiation and progression of breast cancer. RTKs are cell surface receptors with unique structures and biological characteristics, which respond to environmental signals by initiating signaling cascades such as the mitogen-activated protein kinase (MAPK) pathway, Janus kinase (JAK)/signal transducer, activator of transcription (STAT) pathway, and phosphoinositide 3-kinase (PI3K)/AKT pathway. The critical role of RTKs makes them suitable targets for breast cancer treatment. Targeted therapies against RTKs have been developed in recent years, evaluated in clinical trials, and approved for several cancer types, including breast cancer. However, breast cancer displays molecular heterogeneity and exhibits different therapeutic responses to various drug types, leading to limited effectiveness of targeted therapy against RTKs. In this review, we summarize the structural and functional characteristics of selected RTKs and discuss the mechanisms and current status of drug therapy involving different protein tyrosine kinases in breast cancer progression.
Collapse
Affiliation(s)
- Yu Qi
- Department of Pathology, School of Basic Medical Sciences, Central South UniversityChangsha, Hunan, China
| | - Shu-Min Deng
- Department of Pathology, School of Basic Medical Sciences, Central South UniversityChangsha, Hunan, China
| | - Kuan-Song Wang
- Department of Pathology, School of Basic Medical Sciences, Central South UniversityChangsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| |
Collapse
|
4
|
Chen Y, Lu A, Hu Z, Li J, Lu J. ERBB3 targeting: A promising approach to overcoming cancer therapeutic resistance. Cancer Lett 2024; 599:217146. [PMID: 39098760 DOI: 10.1016/j.canlet.2024.217146] [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: 03/26/2024] [Revised: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024]
Abstract
Human epidermal growth factor receptor-3 (ERBB3) is a member of the ERBB receptor tyrosine kinases (RTKs) and is expressed in many malignancies. Along with other ERBB receptors, ERBB3 is associated with regulating normal cell proliferation, apoptosis, differentiation, and survival, and has received increased research attention for its involvement in cancer therapies. ERBB3 expression or co-expression levels have been investigated as predictive factors for cancer prognosis and drug sensitivity. Additionally, the association between the elevated expression of ERBB3 and treatment failure in cancer therapy further established ERBB3-targeting therapy as a crucial therapeutic approach. This review delves into the molecular mechanisms of ERBB3-driven resistance to targeted therapeutics against ERBB2 and EGFR and other signal transduction inhibitors, endocrine therapy, chemotherapy, and radiotherapy. Using preclinical and clinical evidence, we synthesise and explicate how various aspects of aberrant ERBB3 activities-such as compensatory activation, signal crosstalk interactions, dysregulation in the endocytic pathway, mutations, ligand-independent activation, intrinsic kinase activity, and homodimerisation-can lead to resistance development and/or treatment failures. Several ERBB3-directed monoclonal antibodies, bispecific antibodies, and the emerging antibody-drug conjugate demonstrate encouraging clinical outcomes for improving therapeutic efficacy and overcoming resistance, especially when combined with other anti-cancer approaches. More research efforts are needed to identify appropriate biomarkers tailored for ERBB3-targeted therapies.
Collapse
Affiliation(s)
- Yutao Chen
- Auckland Bioengineering Institute, University of Auckland, Auckland, 1142, New Zealand
| | - Anni Lu
- Pinehurst School, Albany, Auckland, New Zealand
| | - Zhangli Hu
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Jinyao Li
- College of Life Sciences, Xijiang University, Urumqi, China
| | - Jun Lu
- Auckland Bioengineering Institute, University of Auckland, Auckland, 1142, New Zealand; College of Food Engineering and Nutrition Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi Province, China; College of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi Province, China; Department of Food and Agriculture Technology, Yangtze Delta Region Institute of Tsinghua University, Zhejiang, Jiaxing, 314006, China.
| |
Collapse
|
5
|
Becker AK, Puladi B, Xie K, Cassataro A, Götzl R, Hölzle F, Beier JP, Knüchel-Clarke R, Braunschweig T. HER3 (ERBB3) amplification in liposarcoma - a putative new therapeutic target? World J Surg Oncol 2024; 22:131. [PMID: 38760830 PMCID: PMC11100077 DOI: 10.1186/s12957-024-03406-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Liposarcomas are among the most common mesenchymal malignancies. However, the therapeutic options are still very limited and so far, targeted therapies had not yet been established. Immunotherapy, which has been a breakthrough in other oncological entities, seems to have no efficacy in liposarcoma. Complicating matters further, classification remains difficult due to the diversity of morphologies and nonspecific or absent markers in immunohistochemistry, leaving molecular pathology using FISH or sequencing as best options. Many liposarcomas harbor MDM2 gene amplifications. In close relation to the gene locus of MDM2, HER3 (ERBB3) gene is present and co-amplification could occur. Since the group of HER/EGFR receptor tyrosine kinases and its inhibitors/antibodies play a role in a broad spectrum of oncological diseases and treatments, and some HER3 inhibitors/antibodies are already under clinical investigation, we hypothesized that in case of HER3 co-amplifications a tumor might bear a further potential therapeutic target. METHODS We performed FISH analysis (MDM2, DDIT3, HER3) in 56 archived cases and subsequently performed reclassification to confirm the diagnosis of liposarcoma. RESULTS Next to 16 out of 56 cases needed to be re-classified, in 20 out of 54 cases, a cluster-amplification of HER3 could be detected, significantly correlating with MDM2 amplification. Our study shows that the entity of liposarcomas show specific molecular characteristics leading to reclassify archived cases by modern, established methodologies. Additionally, in 57.1% of these cases, HER3 was cluster-amplified profusely, presenting a putative therapeutic target for targeted therapy. CONCLUSION Our study serves as the initial basis for further investigation of the HER3 gene as a putative therapeutic target in liposarcoma.
Collapse
Affiliation(s)
| | - Behrus Puladi
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, 52074, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Kunpeng Xie
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, 52074, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Angela Cassataro
- Institute of Pathology, University Hospital RWTH Aachen, 52074, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Rebekka Götzl
- Department of Plastic, Hand Surgery - Burn Center, University Hospital RWTH Aachen, 52074, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Frank Hölzle
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, 52074, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Justus P Beier
- Department of Plastic, Hand Surgery - Burn Center, University Hospital RWTH Aachen, 52074, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Ruth Knüchel-Clarke
- Institute of Pathology, University Hospital RWTH Aachen, 52074, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Till Braunschweig
- Institute of Pathology, University Hospital RWTH Aachen, 52074, Aachen, Germany.
- Institute of Pathology, Faculty of Medicine, LMU Munich, Thalkirchner Strasse 36, 80337, Munich, Germany.
| |
Collapse
|
6
|
Tommasi C, Airò G, Pratticò F, Testi I, Corianò M, Pellegrino B, Denaro N, Demurtas L, Dessì M, Murgia S, Mura G, Wekking D, Scartozzi M, Musolino A, Solinas C. Hormone Receptor-Positive/HER2-Positive Breast Cancer: Hormone Therapy and Anti-HER2 Treatment: An Update on Treatment Strategies. J Clin Med 2024; 13:1873. [PMID: 38610638 PMCID: PMC11012464 DOI: 10.3390/jcm13071873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Hormone receptor (HR)-positive/HER2-positive breast cancer represents a distinct subtype expressing estrogen and progesterone receptors with an overexpression of HER2. Approximately 14% of female breast cancer cases are HER2-positive, with the majority being HR-positive. These tumors show a cross-talk between the hormonal and HER2 pathways; the interaction has implications for the treatment options for the disease. In this review, we analyze the biology of HR-positive/HER2-positive breast cancer and summarize the evidence concerning the standard of care options both in neoadjuvant/adjuvant settings and in advanced disease. Additionally, we focus on new trials and drugs for HR-positive/HER2-positive breast cancer and the new entity: HER2-low breast cancer.
Collapse
Affiliation(s)
- Chiara Tommasi
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Giulia Airò
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Fabiana Pratticò
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Irene Testi
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
| | - Matilde Corianò
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Benedetta Pellegrino
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Nerina Denaro
- Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Laura Demurtas
- Medical Oncology, AOU Cagliari, Policlinico Duilio Casula, 09042 Monserrato, Italy (C.S.)
| | - Mariele Dessì
- Medical Oncology, AOU Cagliari, Policlinico Duilio Casula, 09042 Monserrato, Italy (C.S.)
| | - Sara Murgia
- Medical Oncology, University of Cagliari, 09124 Cagliari, Italy
| | - Giovanni Mura
- Pathological Anatomy, Laboratory Valdès, 81200 Cagliari, Italy
| | - Demi Wekking
- Academic Medical Centre, Amsterdam University Medical Center, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Mario Scartozzi
- Medical Oncology, AOU Cagliari, Policlinico Duilio Casula, 09042 Monserrato, Italy (C.S.)
- Medical Oncology, University of Cagliari, 09124 Cagliari, Italy
| | - Antonino Musolino
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Cinzia Solinas
- Medical Oncology, AOU Cagliari, Policlinico Duilio Casula, 09042 Monserrato, Italy (C.S.)
| |
Collapse
|
7
|
Shao H, Zhao M, Guan AJ, Shao T, Zhou D, Yu G, Tang W. A network meta-analysis of efficacy and safety for first-line and second/further-line therapies in postmenopausal women with hormone receptor-positive, HER2-negative, advanced breast cancer. BMC Med 2024; 22:13. [PMID: 38212842 PMCID: PMC10785354 DOI: 10.1186/s12916-023-03238-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 12/19/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR + /HER2 -) advanced breast cancer is a prevalent subtype among postmenopausal women. Despite the growing number of randomized clinical trials (RCTs) exploring this topic, the efficacy and safety of first-line and second/further-line treatments remain uncertain. Accordingly, our aim was to conduct a comprehensive evaluation of the efficacy and safety of these therapies through network meta-analysis. METHODS RCTs were identified by searching Pubmed, Embase, and major cancer conferences. The efficacy of interventions was assessed using the hazard ratios (HRs) of progression-free survival (PFS) and overall survival (OS), while safety was indicated by the incidence of any grade adverse events (AEs), grade 3-5 AEs, AEs leading to treatment discontinuation, and AEs leading to death. Both time-variant HRs fractional polynomial models and time-invariant HRs Cox-proportional hazards models were considered for handling time-to-event data. Safety indicators were analyzed using Bayesian network meta-analysis. Additionally, subgroup analyses were conducted based on patient characteristics. RESULTS A total of 41 RCTs (first-line 17, second/further-lines 27) were included in the analysis. For first-line treatment, the addition of Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors to endocrine therapy significantly improved therapeutic efficacy in terms of both PFS and OS, demonstrating the best performance across all mechanisms. Specifically, the combination of Abemaciclib and Letrozole demonstrated the most favorable performance in terms of PFS, while Ribociclib plus Fulvestrant yielded the best outcomes in OS. Incorporating the immune checkpoint inhibitor Avelumab into the regimen with CDK4/6 inhibitors and selective estrogen receptor degraders significantly enhanced both PFS and OS in second-line or later treatments. Regarding safety, endocrine monotherapy performed well. Regarding safety, endocrine monotherapy performed well. There is mounting evidence suggesting that most CDK4/6 inhibitors may demonstrate poorer performance with respect to hematologic AEs. However, additional evidence is required to further substantiate these findings. CONCLUSIONS CDK4/6 inhibitors, combined with endocrine therapy, are pivotal in first-line treatment due to their superior efficacy and manageable AEs. For second/further-line treatment, adding immune checkpoint inhibitors to CDK4/6 inhibitors plus endocrine therapy may produce promising results. However, to reduce the results' uncertainty, further trials comparing these novel treatments are warranted. TRIAL REGISTRATION Registration number: PROSPERO (CRD42022377431).
Collapse
Affiliation(s)
- Hanqiao Shao
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
- Center for Pharmacoeconomics and Outcomes Research & Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mingye Zhao
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
- Center for Pharmacoeconomics and Outcomes Research & Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ai-Jia Guan
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Sichuan, China
| | - Taihang Shao
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
- Center for Pharmacoeconomics and Outcomes Research & Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Dachuang Zhou
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
- Center for Pharmacoeconomics and Outcomes Research & Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guo Yu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Wenxi Tang
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China.
- Center for Pharmacoeconomics and Outcomes Research & Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
8
|
Hachey SJ, Hatch CJ, Gaebler D, Mocherla A, Nee K, Kessenbrock K, Hughes CCW. Targeting tumor-stromal interactions in triple-negative breast cancer using a human vascularized micro-tumor model. Breast Cancer Res 2024; 26:5. [PMID: 38183074 PMCID: PMC10768273 DOI: 10.1186/s13058-023-01760-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 12/21/2023] [Indexed: 01/07/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive with limited available treatments. Stromal cells in the tumor microenvironment (TME) are crucial in TNBC progression; however, understanding the molecular basis of stromal cell activation and tumor-stromal crosstalk in TNBC is limited. To investigate therapeutic targets in the TNBC stromal niche, we used an advanced human in vitro microphysiological system called the vascularized micro-tumor (VMT). Using single-cell RNA sequencing, we revealed that normal breast tissue stromal cells activate neoplastic signaling pathways in the TNBC TME. By comparing interactions in VMTs with clinical data, we identified therapeutic targets at the tumor-stromal interface with potential clinical significance. Combining treatments targeting Tie2 signaling with paclitaxel resulted in vessel normalization and increased efficacy of paclitaxel in the TNBC VMT. Dual inhibition of HER3 and Akt also showed efficacy against TNBC. These data demonstrate the potential of inducing a favorable TME as a targeted therapeutic approach in TNBC.
Collapse
Affiliation(s)
- Stephanie J Hachey
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA.
| | | | - Daniela Gaebler
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Aneela Mocherla
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Kevin Nee
- Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Kai Kessenbrock
- Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Christopher C W Hughes
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
9
|
Wang J, Han Y, Wang J, Li Q, Xu B. Endocrine Therapy-Based Strategies for Metastatic Breast Cancer with Different Endocrine Sensitivity Statuses: A Systematic Review and Network Meta-Analysis. Cancers (Basel) 2022; 14:6100. [PMID: 36551586 PMCID: PMC9776369 DOI: 10.3390/cancers14246100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Novel endocrine therapies (ETs) and targeted therapeutic regimens have been developed to dramatically improve the outcome of hormone receptor-positive (HR+)/HER2-negative (HER2-) metastatic breast cancer (mBC). METHODS We performed a systematic search with a predefined search strategy in PubMed, Embase and Cochrane CENTRAL databases to perform a network meta-analysis and evaluate the relative efficacies of ET-based treatment regimens in HR+/HER2- mBC patients with different endocrine sensitivity statuses. The study was registered in the PROSPERO database (CRD42021235570). RESULTS A total of 47 trials (20,267 patients) were included. Analysis of progression-free survival (PFS) in endocrine therapy-sensitive (ETS) patients revealed cyclin-dependent kinases 4/6 inhibitors (CDK4/6i) + fulvestrant 500 mg (Ful 500) (random effect (RE): hazard ratio (HR), 0.46; 95% credibility interval (CrI), 0.27-0.78; surface under the cumulative ranking curve (SUCRA), 0.93; fixed effect (FE): HR, 0.48; 95% CrI, 0.40-0.58; SUCRA, 0.99) to be the best therapy followed by CDK4/6i + aromatase inhibitors (AIs) (RE: HR, 0.53; 95% CrI, 0.40-0.72; SUCRA, 0.86; FE: HR, 0.54; 95% CrI, 0.48-0.61; SUCRA, 0.91). Chemotherapy followed by CDK4/6i + Ful 500 appears to be the most effective option for the endocrine therapy-resistant (ETR) group. Analysis of overall survival revealed CDK4/6i + Ful 500 (SUCRA: 0.99) and AKTi + Ful 500 (SUCRA: 0.87) to be the first-rank regimen for the ETS group and ETR groups, respectively. CONCLUSION Our comprehensive analysis suggests that CDK4/6i combined with ETs may be the best treatment option in terms of PFS for ETS patients and chemotherapy for ETR patients with HR+/HER2- mBC. Different endocrine sensitivity statuses required various optimal treatment strategies, which may provide guidance for clinical practice.
Collapse
Affiliation(s)
- Jiani Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yiqun Han
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jiayu Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qing Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
10
|
Tang Q, Peng T, Hu J, Zhang T, Chen P, Chen D, Wang Y, Chen L, Tong L, Chen Y, Xie H, Liang G. Discovery of N-(3-bromo-1H-indol-5-yl)-quinazolin-4-amine as an effective molecular skeleton to develop reversible/irreversible pan-HER inhibitors. Eur J Med Chem 2022; 233:114249. [DOI: 10.1016/j.ejmech.2022.114249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/24/2022]
|
11
|
Pinzón-García AD, Sinisterra R, Cortes M, Mesa F, Ramírez-Clavijo S. Polycaprolactone nanofibers as an adjuvant strategy for Tamoxifen release and their cytotoxicity on breast cancer cells. PeerJ 2021; 9:e12124. [PMID: 34760343 PMCID: PMC8556714 DOI: 10.7717/peerj.12124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 08/17/2021] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the second leading cause of death in women, and tamoxifen citrate (TMX) is accepted widely for the treatment of hormone receptor-positive breast cancers. Several local drug-delivery systems, including nanofibers, have been developed for antitumor treatment. Nanofibers are biomaterials that mimic the natural extracellular matrix, and they have been used as controlled release devices because they enable highly efficient drug loading. The purpose of the present study was to develop polycaprolactone (PCL) nanofibers incorporating TMX for use in the treatment of breast tumors. Pristine PCL and PCL-TMX nanofibers were produced by electrospinning and characterized physiochemically using different techniques. In addition, an in vitro study of TMX release from the nanofibers was performed. The PCL-TMX nanofibers showed sustained TMX release up to 14 h, releasing 100% of the TMX. The Resazurin reduction assay was used to evaluate the TMX cytotoxicity on MCF-7 breast cancer cell line and PBMCs human. The PCL-TMX nanofiber was cytotoxic toPBMCs and MCF-7. Based on these results, the PCL-TMX nanofibers developed have potential as an alternative for local chronic TMX use for breast cancer treatment, however tissue tests must be done.
Collapse
Affiliation(s)
- Ana D Pinzón-García
- Chemistry Department, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ruben Sinisterra
- Chemistry Department, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Cortes
- Restorative Dentistry Department, Faculty of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fredy Mesa
- Faculty of Natural Sciences, Department of Biology, Universidad del Rosario, Bogotá, Colombia
| | - Sandra Ramírez-Clavijo
- Faculty of Natural Sciences, Department of Biology, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
12
|
Chen S, Ren Y, Dai H, Li Y, Lan B, Ma F. Drug-induced pulmonary toxicity in breast cancer patients treated with systemic therapy: a systematic literature review. Expert Rev Anticancer Ther 2021; 21:1399-1410. [PMID: 34672214 DOI: 10.1080/14737140.2021.1996229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Drug-induced pulmonary toxicity (DIPT) associated with breast cancer (BC) therapy has been a major concern in recent times. DIPT may not be attributed to a single type of therapy because of the concomitant use of other anticancer drugs or along with radiotherapy, which is an independent risk factor for pulmonary toxicity. AREAS COVERED In this systematic literature review, we evaluated the probable cause and prevalence of DIPT in various systemic therapies used in BC treatment. A literature search was conducted in PubMed, Embase and Cochrane database, up to October 2020. Clinical studies reporting DIPT and related clinical manifestations due to systemic therapy in BC treatment were included. A total of 1749 articles were retrieved, and 193 articles were included. EXPERT OPINION : The leading cause of DIPT among patients with BC was targeted therapy followed by chemotherapy containing regimens. A total of 17 studies reported 35 deaths (15 deaths in chemotherapy) due to DIPT. Physicians must take extra precaution while prescribing systemic therapy known to be associated with DIPT and need to be familiar with early diagnosis of DIPT in order to avoid respiratory-related complications during treatment in BC patients.
Collapse
Affiliation(s)
- Shanshan Chen
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Yanhong Ren
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, National Clinical Research Center for Respiratory Disease, Beijing, China
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, National Clinical Research Center for Respiratory Disease, Beijing, China
| | - Yiqun Li
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Bo Lan
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Ma
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Chen J, Wan R, Li Q, Rao Z, Wang Y, Zhang L, Teichmann AT. Utilizing the Hippo pathway as a therapeutic target for combating endocrine-resistant breast cancer. Cancer Cell Int 2021; 21:306. [PMID: 34112175 PMCID: PMC8194146 DOI: 10.1186/s12935-021-01999-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/27/2021] [Indexed: 12/14/2022] Open
Abstract
Drug resistance is always a great obstacle in any endocrine therapy of breast cancer. Although the combination of endocrine therapy and targeted therapy has been shown to significantly improve prognosis, refractory endocrine resistance is still common. Dysregulation of the Hippo pathway is often related to the occurrence and the development of many tumors. Targeted therapies of this pathway have played important roles in the study of triple negative breast cancer (TNBC). Targeting the Hippo pathway in combination with chemotherapy or other targeted therapies has been shown to significantly improve specific antitumor effects and reduce cancer antidrug resistance. Further exploration has shown that the Hippo pathway is closely related to endocrine resistance, and it plays a "co-correlation point" role in numerous pathways involving endocrine resistance, including related pathways in breast cancer stem cells (BCSCs). Agents and miRNAs targeting the components of the Hippo pathway are expected to significantly enhance the sensitivity of breast cancer cells to endocrine therapy. This review initially explains the possible mechanism of the Hippo pathway in combating endocrine resistance, and it concludes by recommending endocrine therapy in combination with therapies targeting the Hippo pathway in the study of endocrine-resistant breast cancers.
Collapse
Affiliation(s)
- Jing Chen
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China.,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Runlan Wan
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Qinqin Li
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China.,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Zhenghuan Rao
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China.,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yanlin Wang
- North Sichuan Medical College, Nanchong, 637000, China
| | - Lei Zhang
- Department of Gynaecology, The Second People's Hospital of Yibin, Yibin, 644000, China
| | - Alexander Tobias Teichmann
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China. .,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
14
|
Fang H, Cavaliere A, Li Z, Huang Y, Marquez-Nostra B. Preclinical Advances in Theranostics for the Different Molecular Subtypes of Breast Cancer. Front Pharmacol 2021; 12:627693. [PMID: 33986665 PMCID: PMC8111013 DOI: 10.3389/fphar.2021.627693] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/22/2021] [Indexed: 12/29/2022] Open
Abstract
Breast cancer is the most common cancer in women worldwide. The heterogeneity of breast cancer and drug resistance to therapies make the diagnosis and treatment difficult. Molecular imaging methods with positron emission tomography (PET) and single-photon emission tomography (SPECT) provide useful tools to diagnose, predict, and monitor the response of therapy, contributing to precision medicine for breast cancer patients. Recently, many efforts have been made to find new targets for breast cancer therapy to overcome resistance to standard of care treatments, giving rise to new therapeutic agents to offer more options for patients with breast cancer. The combination of diagnostic and therapeutic strategies forms the foundation of theranostics. Some of these theranostic agents exhibit high potential to be translated to clinic. In this review, we highlight the most recent advances in theranostics of the different molecular subtypes of breast cancer in preclinical studies.
Collapse
Affiliation(s)
- Hanyi Fang
- PET Center, Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, CT, United States.,Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Alessandra Cavaliere
- PET Center, Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, CT, United States
| | - Ziqi Li
- PET Center, Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, CT, United States.,Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, CT, United States
| | - Bernadette Marquez-Nostra
- PET Center, Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
15
|
Haikala HM, Jänne PA. Thirty Years of HER3: From Basic Biology to Therapeutic Interventions. Clin Cancer Res 2021; 27:3528-3539. [PMID: 33608318 DOI: 10.1158/1078-0432.ccr-20-4465] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/13/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
HER3 is a pseudokinase member of the EGFR family having a role in both tumor progression and drug resistance. Although HER3 was discovered more than 30 years ago, no therapeutic interventions have reached clinical approval to date. Because the evidence of the importance of HER3 is accumulating, increased amounts of preclinical and clinical trials with HER3-targeting agents are emerging. In this review article, we discuss the most recent HER3 biology in tumorigenic events and drug resistance and provide an overview of the current and emerging strategies to target HER3.
Collapse
Affiliation(s)
- Heidi M Haikala
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Brandão M, Maurer C, Ziegelmann PK, Pondé NF, Ferreira A, Martel S, Piccart M, de Azambuja E, Debiasi M, Lambertini M. Endocrine therapy-based treatments in hormone receptor-positive/HER2-negative advanced breast cancer: systematic review and network meta-analysis. ESMO Open 2020; 5:e000842. [PMID: 32847835 PMCID: PMC7451473 DOI: 10.1136/esmoopen-2020-000842] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/15/2020] [Accepted: 06/19/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Several endocrine therapy (ET)-based treatments are available for patients with advanced breast cancer. We assessed the efficacy of different ET-based treatments in patients with hormone receptor-positive/HER2-negative advanced breast cancer with endocrine-sensitive or endocrine-resistant disease. METHODS We searched Medline and Cochrane Central Register of Controlled Trials up to 15 October 2019 and abstracts from major conferences from 2016 to October 2019. We included phase II/III randomised trials, comparing ≥2 ET-based treatments. Progression-free survival (PFS) and overall survival (OS) were analysed by network meta-analyses using MTC Bayesian models based on both fixed-effect and random-effect models; relative treatment effects were measured as HRs and 95% credibility intervals (CrI). All statistical tests were two-sided. Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed and this systematic review is registered in the PROSPERO database. RESULTS 55 publications reporting on 32 trials (n=12 293 patients) were included. Regarding PFS in the endocrine sensitive setting (n=5200; 12 trials), the combination of cyclin-dependent kinases (CDK)4/6-inhibitors (CDK4/6i)+fulvestrant 500 mg (F500) was likely the most effective treatment (surface under the cumulative ranking curve (SUCRA)=97.3%), followed by CDK4/6i+aromatase inhibitor ±goserelin; there was no significant difference between them (HR 0.82; 95% CrI 0.54-1.25). Regarding OS (n=2157; five trials), the most effective treatment was probably CDK4/6i+F500 (SUCRA=97.3%); comparing CDK4/6i+F500 versus F500 held a HR of 0.77 (95% CrI 0.63-0.95). Regarding PFS in the endocrine-resistant setting (n=6635; 20 trials), CDK4/6i+F500 was likely the most effective treatment (SUCRA=95.7%), followed by capivasertib+F500, without significant difference between them (HR 0.91; 95% CrI 0.60-1.36). For OS (n=4377; 11 trials), the most effective treatments were capivasertib+F500 (SUCRA=84.7%) and CDK4/6i+F500 (SUCRA=69.9%). Comparing CDK4/6i+F500 versus F500 held a HR of 0.77 (95% CrI 0.67-0.89). CONCLUSIONS CDK4/6i+F500 is likely the best treatment option in both endocrine-sensitive and endocrine-resistant diseases for PFS, and in endocrine-sensitive patients for OS. Concerning OS in endocrine-resistant patients, capivasertib+F500 and CDK4/6i+F500 are likely the best treatments. PROSPERO REGISTRATION NUMBER CRD42018104628.
Collapse
Affiliation(s)
- Mariana Brandão
- Academic Trials Promoting Team, Institut Jules Bordet, Bruxelles, Belgium
| | - Christian Maurer
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn, University of Cologne, Cologne, Germany
| | - Patricia Klarmann Ziegelmann
- National Institute for Health Technology Assessment (IATS), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Postgraduate Program in Epidemiology and Department of Statistics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Noam F Pondé
- Medical Oncology, AC Camargo Cancer Center, Sao Paulo, Brazil
| | - Arlindo Ferreira
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - Samuel Martel
- Institut Jules Bordet, Bruxelles, Belgium
- CISSS Montérégie-Centre, Hopital Charles-Lemoyne, Greenfield Park, Quebec, Canada
| | - Martine Piccart
- Université Libre de Bruxelles, Institut Jules Bordet, Bruxelles, Belgium
| | - Evandro de Azambuja
- Academic Trials Promoting Team, Institut Jules Bordet, Bruxelles, Belgium
- Department of Medical Oncology, Institut Jules Bordet, Brussels, Belgium
| | | | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C, Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
17
|
Sun ZG, Zhao LH, Li ZN, Zhu HL. Development and Challenges of the Discovery of HER2 Inhibitors. Mini Rev Med Chem 2020; 20:2123-2134. [PMID: 32727326 DOI: 10.2174/1389557520666200729162118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/01/2020] [Accepted: 06/23/2020] [Indexed: 11/22/2022]
Abstract
The treatment of cancer has always been a major problem in the world. Some cancers cannot be treated with surgery, but only with cancer drugs. Among many cancer drugs, small molecule inhibitors play an irreplaceable role. HER2 is one of the HER families, and the development of HER2 inhibitors has made a huge contribution to the treatment of cancer. Some HER2 inhibitors are already on the market, and some HER2 inhibitors are undergoing clinical research. The design, synthesis and development of new HER2 inhibitors targeting different targets are also ongoing, and some are even under clinical research. The HER2 inhibitors that are on the market have developed resistance, which brings great challenges to the HER2 inhibitor development in the future. This article reviews the development and challenges of the discovery of HER2 inhibitors.
Collapse
Affiliation(s)
- Zhi-Gang Sun
- Central Laboratory, Linyi Central Hospital, No.17 Jiankang Road, Linyi 276400, China
| | - Liang-Hui Zhao
- Weifang Medical University, No. 7166 Baotong West Street, Weifang 261000, China
| | - Zhi-Na Li
- Central Laboratory, Linyi Central Hospital, No.17 Jiankang Road, Linyi 276400, China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing 210023, China
| |
Collapse
|
18
|
Kanumuri R, Saravanan R, Pavithra V, Sundaram S, Rayala SK, Venkatraman G. Current trends and opportunities in targeting p21 activated kinase-1(PAK1) for therapeutic management of breast cancers. Gene 2020; 760:144991. [PMID: 32717309 DOI: 10.1016/j.gene.2020.144991] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most frequently diagnosed cancer in women worldwide. Identifying reliable biomarkers and druggable molecular targets pose to be a significant quest in breast cancer research. p21-activated kinase 1 (PAK1) is a serine/threonine kinase that direct cell motility, cytoskeletal remodelling, and has been shown to function as a downstream regulator for various cancer signalling cascades that promote cell proliferation, apoptosis deregulation and hasten mitotic abnormalities, resulting in tumor formation and progression. The heterogeneity and acquired drug resistance are important factors that challenge the treatment of breast cancer. p21-activated kinase 1 signalling is crucial for activation of the Ras/RAF/MEK/ERK, PI3K/Akt/mTOR and Wnt signalling cascades which regulate cell survival, cell cycle progression, differentiation, and proliferation. A study involving proteogenomics analysis on breast cancer tissues showed the PAK1 as outlier kinase. In addition to this, few outlier molecules were identified specific to subtypes of breast cancer. A few substrates of PAK1 in breast cancer are already known. In this paper, we have discussed a similar approach called Kinase Interacting Substrate Screening (KISS) for the identification of novel oncogenic substrates of p21-activated kinase specific to subtypes of breast cancer. Such high throughput approaches are expected to accelerate the process of identifying novel drug targets and biomarkers.
Collapse
Affiliation(s)
- Rahul Kanumuri
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India; Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - Roshni Saravanan
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - V Pavithra
- Department of Pathology, Sri Ramachandra Medical College & Research Institute, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - Sandhya Sundaram
- Department of Pathology, Sri Ramachandra Medical College & Research Institute, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - Suresh K Rayala
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, India.
| | - Ganesh Venkatraman
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India.
| |
Collapse
|
19
|
Zhou J, Le K, Xu M, Ming J, Yang W, Zhang Q, Lu L, Xi Z, Ruan S, Huang T. CXCR4 Antagonist AMD3100 Reverses the Resistance to Tamoxifen in Breast Cancer via Inhibiting AKT Phosphorylation. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:161-170. [PMID: 32691010 PMCID: PMC7311345 DOI: 10.1016/j.omto.2020.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 06/19/2020] [Indexed: 01/11/2023]
Abstract
Endocrine therapy is a systemic therapy and has become the main treatment strategy for patients with estrogen receptor (ER)-positive breast cancer. However, tamoxifen resistance has become an insurmountable clinical challenge, and the underlying mechanisms are still poorly understood. In this study, we explored the roles of CXC chemokine receptor type 4 (CXCR4) in tamoxifen-treated breast cancer and tamoxifen resistance. Based on the Gene Expression Omnibus (GEO) database, high expression of CXCR4 was found to be associated with worse overall survival (hazard ratio [HR] = 4.646, p < 0.001) and cancer-specific survival (HR = 4.480, p < 0.001) in tamoxifen-treated breast cancer. CXCR4 was also positively correlated with the level of AKT phosphorylation and the resistance to tamoxifen in breast cancer. AMD3100 is a CXCR4 antagonist and was found to decrease phosphorylated (p)-AKT levels of tamoxifen-resistant cells. The reversal effect of AMD3100 on tamoxifen resistance was also confirmed in vitro and in vivo. Taken together, our study demonstrated that CXCR4 could be a potential prognostic biomarker for tamoxifen-treated breast cancer, and the combination of AMD3100 with tamoxifen could be a more efficacious therapeutic strategy for the treatment of tamoxifen resistance.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kehao Le
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ming Xu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jie Ming
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wen Yang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qiulei Zhang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Linlin Lu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zihan Xi
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shengnan Ruan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
20
|
Peters MF, Landry T, Pin C, Maratea K, Dick C, Wagoner MP, Choy AL, Barthlow H, Snow D, Stevens Z, Armento A, Scott CW, Ayehunie S. Human 3D Gastrointestinal Microtissue Barrier Function As a Predictor of Drug-Induced Diarrhea. Toxicol Sci 2020; 168:3-17. [PMID: 30364994 PMCID: PMC6390652 DOI: 10.1093/toxsci/kfy268] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Drug-induced gastrointestinal toxicities (GITs) rank among the most common clinical side effects. Preclinical efforts to reduce incidence are limited by inadequate predictivity of in vitro assays. Recent breakthroughs in in vitro culture methods support intestinal stem cell maintenance and continual differentiation into the epithelial cell types resident in the intestine. These diverse cells self-assemble into microtissues with in vivo-like architecture. Here, we evaluate human GI microtissues grown in transwell plates that allow apical and/or basolateral drug treatment and 96-well throughput. Evaluation of assay utility focused on predictivity for diarrhea because this adverse effect correlates with intestinal barrier dysfunction which can be measured in GI microtissues using transepithelial electrical resistance (TEER). A validation set of widely prescribed drugs was assembled and tested for effects on TEER. When the resulting TEER inhibition potencies were adjusted for clinical exposure, a threshold was identified that distinguished drugs that induced clinical diarrhea from those that lack this liability. Microtissue TEER assay predictivity was further challenged with a smaller set of drugs whose clinical development was limited by diarrhea that was unexpected based on 1-month animal studies. Microtissue TEER accurately predicted diarrhea for each of these drugs. The label-free nature of TEER enabled repeated quantitation with sufficient precision to develop a mathematical model describing the temporal dynamics of barrier damage and recovery. This human 3D GI microtissue is the first in vitro assay with validated predictivity for diarrhea-inducing drugs. It should provide a platform for lead optimization and offers potential for dose schedule exploration.
Collapse
Affiliation(s)
- Matthew F Peters
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Tim Landry
- MatTek Corporation, Ashland, Massachusetts 01721
| | - Carmen Pin
- Mechanistic Safety and ADME Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, CB4 0WG, UK
| | - Kim Maratea
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Cortni Dick
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Matthew P Wagoner
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Allison L Choy
- Science and Enabling Units IT, AstraZeneca, Waltham, MA 02451
| | - Herb Barthlow
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Deb Snow
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | | | - Alex Armento
- MatTek Corporation, Ashland, Massachusetts 01721
| | - Clay W Scott
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | | |
Collapse
|
21
|
Thomas A, Virdee PS, Eatock M, Lord SR, Falk S, Anthoney DA, Turkington RC, Goff M, Elhussein L, Collins L, Love S, Moschandreas J, Middleton MR. Dual Erb B Inhibition in Oesophago-gastric Cancer (DEBIOC): A phase I dose escalating safety study and randomised dose expansion of AZD8931 in combination with oxaliplatin and capecitabine chemotherapy in patients with oesophagogastric adenocarcinoma. Eur J Cancer 2020; 124:131-141. [PMID: 31765988 PMCID: PMC6947485 DOI: 10.1016/j.ejca.2019.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/07/2019] [Accepted: 10/13/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND AZD8931 has equipotent activity against epidermal growth factor receptor, erbB2, and erbB3. Primary objectives were to determine the recommended phase II dose (RP2D) of AZD8931 + chemotherapy, and subsequently assess safety/preliminary clinical activity in patients with operable oesophagogastric cancer (OGC). METHODS AZD8931 (20 mg, 40 mg or 60 mg bd) was given with Xelox (oxaliplatin + capecitabine) for eight 21-day cycles, continuously or with intermittent schedule (4 days on/3 off every week; 14 days on/7 off, per cycle) in a rolling-six design. Subsequently, patients with OGC were randomised 2:1 to AZD8931 + Xelox at RP2D or Xelox only for two cycles, followed by radical oesophagogastric surgery. Secondary outcomes were safety, complete resection (R0) rate, six-month progression-free survival (PFS) and overall survival. RESULTS During escalation, four dose-limiting toxicities were observed among 24 patients: skin rash (1) and failure to deliver 100% of Xelox because of treatment-associated grade III-IV adverse events (AEs) (3: diarrhoea and vomiting; vomiting; fatigue). Serious adverse events (SAE) occurred in 15 of 24 (63%) patients. RP2D was 20-mg bd with the 4/3 schedule. In the expansion phase, 2 of 20 (10%) patients in the Xelox + AZD8931 group and 5/10 (50%) patients in the Xelox group had grade III-IV AEs. Six-month PFS was 85% (90% CI: 66%-94%) in Xelox + AZD8931 and 100% in Xelox alone. Seven deaths (35%) occurred with Xelox + AZD8931 and one (10%) with Xelox. R0 rate was 45% (9/20) with Xelox + AZD8931 and 90% (9/10) with Xelox-alone (P = 0.024). CONCLUSION Xelox + AZD8931 (20 mg bd 4/3 days) has an acceptable safety profile administered as neoadjuvant therapy in operable patients with OGC. (Trial registration: EudraCT 2011-003169-13, ISRCTN-68093791).
Collapse
Affiliation(s)
| | - Pradeep S Virdee
- Centre for Statistics in Medicine, University of Oxford, Oxford, UK
| | | | | | - Stephen Falk
- Bristol Haematology & Oncology Centre, Bristol, UK
| | | | - Richard C Turkington
- Centre for Cancer Research and Cell Biology, Queens University Belfast, Belfast, UK
| | - Matthew Goff
- Oncology Clinical Trials Office, University of Oxford, Oxford, UK
| | - Leena Elhussein
- Centre for Statistics in Medicine, University of Oxford, Oxford, UK
| | - Linda Collins
- Oncology Clinical Trials Office, University of Oxford, Oxford, UK
| | - Sharon Love
- Centre for Statistics in Medicine, University of Oxford, Oxford, UK
| | | | - Mark R Middleton
- University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, UK
| |
Collapse
|
22
|
Montagna E, Colleoni M. Hormonal treatment combined with targeted therapies in endocrine-responsive and HER2-positive metastatic breast cancer. Ther Adv Med Oncol 2019; 11:1758835919894105. [PMID: 31897091 PMCID: PMC6918494 DOI: 10.1177/1758835919894105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 09/04/2019] [Indexed: 01/03/2023] Open
Abstract
Approximately 50% of HER2 positive breast cancer cases are also estrogen receptor (ER) positive. Data supports a role for close cross-talk between the ER and HER2 signaling pathways as an important contributor to the development of de novo or acquired resistance to hormone therapies. Therefore a strategy that simultaneously blocks both signaling pathways is a reasonable approach to prevent or overcome either endocrine or anti-HER2 therapy resistance. Moreover, preclinical data support the idea that PI3K inhibitors and CDK4/6 could be an attractive target that functions downstream of both ER and HER2 pathways. We conducted a literature review of the results of phase II and III studies testing targeted therapies in metastatic breast cancer with HER2-positive and hormonal-receptor-positive disease. The analyses included efficacy and toxicity data from earlier studies with a single anti-HER2 drug combined with hormonal therapy up to more recent studies testing new molecules targeting these signaling pathways. The aims of this review are to summarize current knowledge and to discuss research development including the possibility to spare chemotherapy in this subgroup of HER2-positive breast cancer patients.
Collapse
Affiliation(s)
- Emilia Montagna
- Division of Medical Senology, European Institute of Oncology, Via Ripamonti 435, Milan, 20141, Italy
| | - Marco Colleoni
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
23
|
Aggelis V, Johnston SRD. Advances in Endocrine-Based Therapies for Estrogen Receptor-Positive Metastatic Breast Cancer. Drugs 2019; 79:1849-1866. [DOI: 10.1007/s40265-019-01208-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
24
|
Wang M, Hu Y, Yu T, Ma X, Wei X, Wei Y. Pan-HER-targeted approach for cancer therapy: Mechanisms, recent advances and clinical prospect. Cancer Lett 2018; 439:113-130. [PMID: 30218688 DOI: 10.1016/j.canlet.2018.07.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/08/2018] [Accepted: 07/09/2018] [Indexed: 02/05/2023]
Abstract
The Human Epidermal Growth Factor Receptor family is composed of 4 structurally related receptor tyrosine kinases that are involved in many human cancers. The efficacy and safety of HER inhibitors have been compared in a wide range of clinical trials, suggesting the superior inhibitory ability of multiple- HER-targeting blockade compared with single receptor antagonists. However, many patients are currently resistant to current therapeutic treatment and novel strategies are warranted to conquer the resistance. Thus, we performed a critical review to summarize the molecular involvement of HER family receptors in tumour progression, recent anti-HER drug development based on clinical trials, and the potential resistance mechanisms of anti-HER therapy.
Collapse
Affiliation(s)
- Manni Wang
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Yuzhu Hu
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Ting Yu
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Xuelei Ma
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Xiawei Wei
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China.
| | - Yuquan Wei
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| |
Collapse
|
25
|
Yau THL, Cheung KL. Optimising endocrine therapy in postmenopausal women with advanced breast cancer. Endocr Relat Cancer 2018; 25:705-721. [PMID: 29674428 DOI: 10.1530/erc-18-0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 04/19/2018] [Indexed: 01/01/2023]
Abstract
Hormone receptor-positive breast cancer is commonly treated with endocrine therapy (ET); however, over time, cancer cells can develop endocrine resistance. This review aims to document combination therapy and sequential therapy in the use of endocrine agents and targeted agents, by conducting two systematic searches using four databases: Cochrane Library, MEDLINE, EMBASE and Web of Science. A total of 26 studies that covered combination therapy were obtained and included for the review. Fourteen were phase III documenting combinations of mechanistic target of rapamycin (mTOR), phosphoinositide-3-kinase (PI3K), vascular endothelial growth factor receptor, human epidermal growth factor receptor 2 and cyclin-dependent kinase 4/6 (CDK4/6) inhibitors. The remaining studies were of phase II nature that reported combinations involving inhibitors in mTOR, endothelial growth factor receptor, CDK4/6 and TKI. Interesting findings in inhibitor combinations involving CDK4/6, mTOR and PI3K suggest clinical activity that can overcome endocrine resistance. On the other hand, there were 0 studies that covered sequential therapy. Overall findings showed that combination therapy improved treatment efficacy over monotherapy in postmenopausal patients with hormone receptor-positive advanced breast cancer. Inevitably, the benefits are accompanied with increased toxicity. To optimise ET, further research into combinations and effective patient selection will need to be defined. Additionally, this review warrants future studies to explore sequential therapy.
Collapse
|
26
|
Ribas R, Pancholi S, Rani A, Schuster E, Guest SK, Nikitorowicz-Buniak J, Simigdala N, Thornhill A, Avogadri-Connors F, Cutler RE, Lalani AS, Dowsett M, Johnston SR, Martin LA. Targeting tumour re-wiring by triple blockade of mTORC1, epidermal growth factor, and oestrogen receptor signalling pathways in endocrine-resistant breast cancer. Breast Cancer Res 2018; 20:44. [PMID: 29880014 PMCID: PMC5992820 DOI: 10.1186/s13058-018-0983-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/10/2018] [Indexed: 12/19/2022] Open
Abstract
Background Endocrine therapies are the mainstay of treatment for oestrogen receptor (ER)-positive (ER+) breast cancer (BC). However, resistance remains problematic largely due to enhanced cross-talk between ER and growth factor pathways, circumventing the need for steroid hormones. Previously, we reported the anti-proliferative effect of everolimus (RAD001-mTORC1 inhibitor) with endocrine therapy in resistance models; however, potential routes of escape from treatment via ERBB2/3 signalling were observed. We hypothesised that combined targeting of three cellular nodes (ER, ERBB, and mTORC1) may provide enhanced long-term clinical utility. Methods A panel of ER+ BC cell lines adapted to long-term oestrogen deprivation (LTED) and expressing ESR1wt or ESR1Y537S, modelling acquired resistance to an aromatase-inhibitor (AI), were treated in vitro with a combination of RAD001 and neratinib (pan-ERBB inhibitor) in the presence or absence of oestradiol (E2), tamoxifen (4-OHT), or fulvestrant (ICI182780). End points included proliferation, cell signalling, cell cycle, and effect on ER-mediated transactivation. An in-vivo model of AI resistance was treated with monotherapies and combinations to assess the efficacy in delaying tumour progression. RNA-seq analysis was performed to identify changes in global gene expression as a result of the indicated therapies. Results Here, we show RAD001 and neratinib (pan-ERBB inhibitor) caused a concentration-dependent decrease in proliferation, irrespective of the ESR1 mutation status. The combination of either agent with endocrine therapy further reduced proliferation but the maximum effect was observed with a triple combination of RAD001, neratinib, and endocrine therapy. In the absence of oestrogen, RAD001 caused a reduction in ER-mediated transcription in the majority of the cell lines, which associated with a decrease in recruitment of ER to an oestrogen-response element on the TFF1 promoter. Contrastingly, neratinib increased both ER-mediated transactivation and ER recruitment, an effect reduced by the addition of RAD001. In-vivo analysis of an LTED model showed the triple combination of RAD001, neratinib, and fulvestrant was most effective at reducing tumour volume. Gene set enrichment analysis revealed that the addition of neratinib negated the epidermal growth factor (EGF)/EGF receptor feedback loops associated with RAD001. Conclusions Our data support the combination of therapies targeting ERBB2/3 and mTORC1 signalling, together with fulvestrant, in patients who relapse on endocrine therapy and retain a functional ER. Electronic supplementary material The online version of this article (10.1186/s13058-018-0983-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ricardo Ribas
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Sunil Pancholi
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Aradhana Rani
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Eugene Schuster
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Stephanie K Guest
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Joanna Nikitorowicz-Buniak
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Nikiana Simigdala
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Allan Thornhill
- Centre for Cancer Imaging, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | | | | | | | - Mitch Dowsett
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK.,The Ralph Lauren Centre for Breast Cancer Research, The Royal Marsden Hospital, London, SW3 6JJ, UK
| | | | - Lesley-Ann Martin
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW7 3RP, UK.
| |
Collapse
|
27
|
Adams R, Brown E, Brown L, Butler R, Falk S, Fisher D, Kaplan R, Quirke P, Richman S, Samuel L, Seligmann J, Seymour M, Shiu KK, Wasan H, Wilson R, Maughan T. Inhibition of EGFR, HER2, and HER3 signalling in patients with colorectal cancer wild-type for BRAF, PIK3CA, KRAS, and NRAS (FOCUS4-D): a phase 2-3 randomised trial. Lancet Gastroenterol Hepatol 2018; 3:162-171. [PMID: 29254887 PMCID: PMC6125825 DOI: 10.1016/s2468-1253(17)30394-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/25/2017] [Accepted: 11/09/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND A substantial change in trial methodology for solid tumours has taken place, in response to increased understanding of cancer biology. FOCUS4 is a phase 2-3 trial programme testing targeted agents in patients with advanced colorectal cancer in molecularly stratified cohorts. Here, we aimed to test the hypothesis that combined inhibition of EGFR, HER2, and HER3 signalling with the tyrosine kinase inhibitor AZD8931 will control growth of all wild-type tumours. METHODS In FOCUS4-D, we included patients from 18 hospitals in the UK with newly diagnosed advanced or metastatic colorectal cancer whose tumour was wild-type for BRAF, PIK3CA, KRAS, and NRAS. After 16 weeks of first-line therapy, patients with stable or responding tumours were randomised to oral AZD8931 (40 mg twice a day) or placebo. Randomisation was done by minimisation with a random element of 20%, minimisation by hospital site, site of primary tumour, WHO performance status, 16-week CT scan result, number of metastatic sites, and first-line chemotherapy regimen. The primary outcome was progression-free-survival. CT scans were assessed by local radiologists according to Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1. Preplanned interim analyses were assessed per protocol and were agreed using multiarm multistage (MAMS) trial design methodology triggered by occurrence of progression-free survival events in the placebo group. The final analysis was assessed by intention to treat. This trial is registered at controlled-trials.com, ISRCTN 90061546. FINDINGS Between July 7, 2014, and March 7, 2016, 32 patients were randomised to study treatment, 16 to AZD8931 and 16 to placebo. At the first preplanned interim analysis (March, 2016), the independent data monitoring committee (IDMC) recommended closure of FOCUS4-D because of a lack of activity. At the final analysis (Aug 1, 2016), 31 patients had had a progression-free survival event (15 with AZD8931 and 16 with placebo). Median progression-free survival was 3·48 months (95% CI 1·51-5·09) in the placebo group and 2·96 months (1·94-5·62) in the AZD8931 group. No progression-free survival benefit of AZD8931 compared with placebo was noted (hazard ratio [HR] 1·10, 95% CI 0·47-3·57; p=0·95). The most common grade 3 adverse event in the AZD8931 group was skin rash (three [20%] of 15 patients with available data vs none of 16 patients in the placebo group), and in the placebo group it was diarrhoea (one [7%] vs one [6%]). No grade 4 adverse events were recorded and no treatment-related deaths were reported. INTERPRETATION The MAMS trial design for FOCUS4 has shown efficiency and effectiveness in trial outcome delivery, informing the decision to proceed or stop clinical evaluation of a targeted treatment within a molecularly defined cohort of patients. The overarching FOCUS4 trial is now aiming to open a replacement arm in the cohort with all wild-type tumours. FUNDING Medical Research Council (MRC) and National Institute for Health Research (NIHR) Efficacy and Mechanism Evaluation programme, Cancer Research UK, NIHR Clinical Trials Research Network, Health and Care Research Wales, and AstraZeneca.
Collapse
Affiliation(s)
- Richard Adams
- Cardiff University and Velindre Cancer Centre, Cardiff, UK.
| | - Ewan Brown
- Edinburgh Cancer Centre NHS Lothian, Edinburgh, UK
| | - Louise Brown
- Medical Research Council (MRC) Clinical Trials Unit at University College London (UCL), London, UK.
| | - Rachel Butler
- All Wales Genetics Laboratory University Hospital of Wales, Cardiff, UK
| | - Stephen Falk
- University Hospital Bristol NHS Foundation Trust, Bristol, UK
| | - David Fisher
- Medical Research Council (MRC) Clinical Trials Unit at University College London (UCL), London, UK
| | - Richard Kaplan
- Medical Research Council (MRC) Clinical Trials Unit at University College London (UCL), London, UK
| | - Phil Quirke
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Susan Richman
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | | | - Jenny Seligmann
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Matt Seymour
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | | | | | - Richard Wilson
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Tim Maughan
- Cancer Research UK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
28
|
Reinert T, Debiasi M, Bines J, Barrios CH. Trends in progression-free survival (PFS) and time to progression (TTP) over time within first-line aromatase inhibitors trials in hormone receptor-positive advanced breast cancer. Breast Cancer Res Treat 2017; 168:457-465. [PMID: 29190006 DOI: 10.1007/s10549-017-4593-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/22/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Over the last 20 years, aromatase inhibitors (AI) have been tested in clinical trials as first-line therapy for hormone receptor-positive (HR-positive) advanced breast cancer (ABC), firstly as experimental arms, when they proved to be effective, and recently as control arms. This analysis aims to evaluate trends in progression-free survival (PFS) and time to progression (TTP) over time. PATIENTS AND METHODS A literature review was conducted using the MEDLINE database to identify randomized controlled phase II or III trials which reported PFS or TTP of at least one arm using first-line AI HR-positive ABC patients. A linear correlation was used to access the association between the year of the first patient enrolled and the observed PFS/TTP. RESULTS The search retrieved 19 trials, accounting for 4552 postmenopausal patients divided into 21 separate AI treatment arms. The PFS/TTP increased from 6 to 9 months in the earlier trials to 13-16 months in the current era, representing an absolute gain of approximately 7 months, without the addition of any other drug. Our analysis showed a positive correlation between the year of the first patient enrolled in these trials and median PFS/TTP reported (R 2 = 0.34; p < 0.01). No correlation was found between the year of the first patient included in these trials and other potential prognostic factors such as visceral metastasis at baseline (R 2 = 0.26; p = 0.20) or exposure to adjuvant therapy (R 2 = 0.05; p = 0.18). CONCLUSION Patients treated with first-line AIs in the more recently conducted trials have longer PFS/TTP when compared to their counterparts treated with the same drugs in older studies. These findings have important implications for the estimation of sample size and follow-up periods for the planning of future trials as well as in the translation of the results into clinical practice decisions.
Collapse
Affiliation(s)
- T Reinert
- Latin American Cooperative Oncology Group (LACOG), Porto Alegre, Brazil.,Hospital Do Câncer Mãe de Deus, Rua Costa, 40, 4 Andar, Porto Alegre, Brazil.,Post Graduate Program in Medical Sciences, Federal University of Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil
| | - M Debiasi
- Latin American Cooperative Oncology Group (LACOG), Porto Alegre, Brazil.,PUCRS School of Medicine, Porto Alegre, Brazil
| | - J Bines
- Latin American Cooperative Oncology Group (LACOG), Porto Alegre, Brazil.,Instituto Nacional Do Câncer José de Alencar Gomes Da Silva (INCA), Rio De Janeiro, Brazil
| | - C H Barrios
- Latin American Cooperative Oncology Group (LACOG), Porto Alegre, Brazil. .,Hospital Do Câncer Mãe de Deus, Rua Costa, 40, 4 Andar, Porto Alegre, Brazil.
| |
Collapse
|
29
|
Maurer C, Martel S, Zardavas D, Ignatiadis M. New agents for endocrine resistance in breast cancer. Breast 2017; 34:1-11. [DOI: 10.1016/j.breast.2017.04.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/14/2017] [Accepted: 04/16/2017] [Indexed: 11/25/2022] Open
|
30
|
Guerrero-Zotano AL, Arteaga CL. Neoadjuvant Trials in ER + Breast Cancer: A Tool for Acceleration of Drug Development and Discovery. Cancer Discov 2017; 7:561-574. [PMID: 28495849 PMCID: PMC5497752 DOI: 10.1158/2159-8290.cd-17-0228] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/03/2017] [Accepted: 04/17/2017] [Indexed: 12/17/2022]
Abstract
Neoadjuvant therapy trials offer an excellent strategy for drug development and discovery in breast cancer, particularly in triple-negative and HER2-overexpressing subtypes, where pathologic complete response is a good surrogate of long-term patient benefit. For estrogen receptor-positive (ER+) breast cancers, however, use of this strategy has been challenging because of the lack of validated surrogates of long-term efficacy and the overall good prognosis of the majority of patients with this cancer subtype. We review below the clinical benefits of neoadjuvant endocrine therapy for ER+/HER2-negative breast cancer, its use and limitations for drug development, prioritization of adjuvant and metastatic trials, and biomarker discovery.Significance: Neoadjuvant endocrine therapy is an excellent platform for the development of investigational drugs, triaging of novel combinations, biomarker validation, and discovery of mechanisms of drug resistance. This review summarizes the clinical and investigational benefits of this approach, with a focus on how to best integrate predictive biomarkers into novel clinical trial designs. Cancer Discov; 7(6); 561-74. ©2017 AACR.
Collapse
Affiliation(s)
- Angel L Guerrero-Zotano
- Departments of Medicine and Cancer Biology; Breast Cancer Program, Vanderbilt-Ingram Cancer Center; Vanderbilt University Medical Center, Nashville, Tennessee
| | - Carlos L Arteaga
- Departments of Medicine and Cancer Biology; Breast Cancer Program, Vanderbilt-Ingram Cancer Center; Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|