1
|
Hickish T, Mehta A, Liu MC, Huang CS, Arora RS, Chang YC, Yang Y, Vladimirov V, Jain M, Tsang J, Pemberton K, Sadrolhefazi B, Jin X, Tseng LM. Afatinib alone and in combination with vinorelbine or paclitaxel, in patients with HER2-positive breast cancer who failed or progressed on prior trastuzumab and/or lapatinib (LUX-Breast 2): an open-label, multicenter, phase II trial. Breast Cancer Res Treat 2022; 192:593-602. [PMID: 35138529 PMCID: PMC8960620 DOI: 10.1007/s10549-021-06449-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE Resistance to HER2 (ErbB2)-targeted therapy may be mediated by other members of the ErbB family. We investigated the efficacy and safety of the irreversible ErbB family blocker, afatinib, alone as first-line therapy in the advanced setting and in combination with vinorelbine or paclitaxel for those who progressed on afatinib monotherapy, in female patients with metastatic breast cancer who had failed or progressed on prior HER2-targeted therapy in the early disease setting. METHODS In this phase II, single-arm, two-part study (ClinicalTrials.gov: NCT01271725), patients in part A received afatinib 40 mg/day in 21-day cycles until disease progression or intolerable adverse events (AEs). Patients with progressive disease could then receive afatinib plus weekly vinorelbine 25 mg/m2 or paclitaxel 80 mg/m2 until disease progression or intolerable AEs (part B). The primary endpoint was confirmed objective response rate (RECIST v1.1). RESULTS Eighty-seven patients were enrolled and 74 were treated in part A (median age: 51 years [range 27-76]; 31 [42%] estrogen receptor-positive, 26 [35%] progesterone receptor-positive). Of these, 39 (53%) patients went on to receive afatinib plus vinorelbine (13 patients) or paclitaxel (26 patients) in part B. Thirteen (18%) and 12 (31%) patients achieved an objective response in parts A and B, respectively. The most common treatment-related AEs with afatinib monotherapy (any/grade ≥ 3) were diarrhea (68%/8%) and rash (49%/4%). Combination therapy was generally well tolerated, with no additive toxicity observed. CONCLUSION Afatinib treatment, alone or in combination with vinorelbine or paclitaxel, was associated with objective responses in ≥ 18% of patients with metastatic breast cancer for whom prior HER2-targeted therapy has failed. Treatment-related AEs were generally manageable, with few grade ≥ 3 AEs reported. TRIAL REGISTRATION ClinicalTrials.gov, NCT01271725, registered 1 July 2011.
Collapse
Affiliation(s)
- Tamas Hickish
- Royal Bournemouth Hospital/Bournemouth University, Castle Ln E, Bournemouth, BH7 7DW, UK.
| | - Ajay Mehta
- Central India Cancer Research Institute, Nagpur, India
| | - Mei-Ching Liu
- Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | | | | | | | - Youngsen Yang
- Division of Hematology-Oncology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Internal Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | | | | | - Janice Tsang
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | | | | | - Xidong Jin
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | | |
Collapse
|
2
|
Chen Y, Zhu S, Pei Y, Hu J, Hu Z, Liu X, Wang X, Gu M, Hu S, Liu X. Differential microRNA Expression in Newcastle Disease Virus-Infected HeLa Cells and Its Role in Regulating Virus Replication. Front Oncol 2021; 11:616809. [PMID: 34150610 PMCID: PMC8211993 DOI: 10.3389/fonc.2021.616809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
As an oncolytic virus, Newcastle disease virus (NDV) can specifically kill tumor cells and has been tested as an attractive oncolytic agent for cancer virotherapy. Virus infection can trigger the changes of the cellular microRNA (miRNA) expression profile, which can greatly influence viral replication and pathogenesis. However, the interplay between NDV replication and cellular miRNA expression in tumor cells is still largely unknown. In the present study, we compared the profiles of cellular miRNAs in uninfected and NDV-infected HeLa cells by small RNA deep sequencing. Here we report that NDV infection in HeLa cells significantly changed the levels of 40 miRNAs at 6 h post-infection (hpi) and 62 miRNAs at 12 hpi. Among 23 highly differentially expressed miRNAs, NDV infection greatly promoted the levels of 3 miRNAs and suppressed the levels of 20 miRNAs at both time points. These 23 miRNAs are predicted to target various genes involved in virus replication and antiviral immunity such as ErbB, Jak-STAT, NF-kB and RIG-I-like receptor. Verification of deep sequencing results by quantitative RT-PCR showed that 9 out of 10 randomly selected miRNAs chosen from this 23-miRNA pool were consistent with deep sequencing data, including 6 down-regulated and 3 up-regulated. Further functional research revealed that hsa-miR-4521, a constituent in this 23-miRNA pool, inhibited NDV replication in HeLa cells. Moreover, dual-luciferase and gene expression array uncovered that the member A of family with sequence similarity 129 (FAM129A) was directly targeted by hsa-miR-4521 and positively regulated NDV replication in HeLa cells, indicating that hsa-miR-4521 may regulate NDV replication via interaction with FAM129A. To our knowledge, this is the first report of the dynamic cellular miRNA expression profile in tumor cells after NDV infection and may provide a valuable basis for further investigation on the roles of miRNAs in NDV-mediated oncolysis.
Collapse
Affiliation(s)
- Yu Chen
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Shanshan Zhu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yuru Pei
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Jiao Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Zenglei Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Xiaowen Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Min Gu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| |
Collapse
|
3
|
Collins DM, Madden SF, Gaynor N, AlSultan D, Le Gal M, Eustace AJ, Gately KA, Hughes C, Davies AM, Mahgoub T, Ballot J, Toomey S, O'Connor DP, Gallagher WM, Holmes FA, Espina V, Liotta L, Hennessy BT, O'Byrne KJ, Hasmann M, Bossenmaier B, O'Donovan N, Crown J. Effects of HER Family-targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer. Clin Cancer Res 2020; 27:807-818. [PMID: 33122343 DOI: 10.1158/1078-0432.ccr-20-2007] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/18/2020] [Accepted: 10/22/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Antibody-dependent cell-mediated cytotoxicity (ADCC) is one mechanism of action of the monoclonal antibody (mAb) therapies trastuzumab and pertuzumab. Tyrosine kinase inhibitors (TKIs), like lapatinib, may have added therapeutic value in combination with mAbs through enhanced ADCC activity. Using clinical data, we examined the impact of lapatinib on HER2/EGFR expression levels and natural killer (NK) cell gene signatures. We investigated the ability of three TKIs (lapatinib, afatinib, and neratinib) to alter HER2/immune-related protein levels in preclinical models of HER2-positive (HER2+) and HER2-low breast cancer, and the subsequent effects on trastuzumab/pertuzumab-mediated ADCC. EXPERIMENTAL DESIGN Preclinical studies (proliferation assays, Western blotting, high content analysis, and flow cytometry) employed HER2+ (SKBR3 and HCC1954) and HER2-low (MCF-7, T47D, CAMA-1, and CAL-51) breast cancer cell lines. NCT00524303 provided reverse phase protein array-determined protein levels of HER2/pHER2/EGFR/pEGFR. RNA-based NK cell gene signatures (CIBERSORT/MCP-counter) post-neoadjuvant anti-HER2 therapy were assessed (NCT00769470/NCT01485926). ADCC assays utilized flow cytometry-based protocols. RESULTS Lapatinib significantly increased membrane HER2 levels, while afatinib and neratinib significantly decreased levels in all preclinical models. Single-agent lapatinib increased HER2 or EGFR levels in 10 of 11 (91%) tumor samples. NK cell signatures increased posttherapy (P = 0.03) and associated with trastuzumab response (P = 0.01). TKI treatment altered mAb-induced NK cell-mediated ADCC in vitro, but it did not consistently correlate with HER2 expression in HER2+ or HER2-low models. The ADCC response to trastuzumab and pertuzumab combined did not exceed either mAb alone. CONCLUSIONS TKIs differentially alter tumor cell phenotype which can impact NK cell-mediated response to coadministered antibody therapies. mAb-induced ADCC response is relevant when rationalizing combinations for clinical investigation.
Collapse
Affiliation(s)
- Denis M Collins
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland.
| | - Stephen F Madden
- RCSI Division of Population Health Sciences, Royal College of Surgeons in Ireland, Beaux Lane House, Dublin, Ireland
| | - Nicola Gaynor
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland
| | - Dalal AlSultan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland.,RCSI Division of Population Health Sciences, Royal College of Surgeons in Ireland, Beaux Lane House, Dublin, Ireland
| | - Marion Le Gal
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland
| | - Alex J Eustace
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland
| | - Kathy A Gately
- Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| | - Clare Hughes
- Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| | - Anthony M Davies
- Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| | - Thamir Mahgoub
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland
| | - Jo Ballot
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Sinead Toomey
- RCSI Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Education & Research Centre, Beaumont Hospital, Beaumont, Dublin, Ireland
| | - Darran P O'Connor
- Royal College of Surgeons in Ireland, School of Pharmacy & Biomolecular Science, Dublin, Ireland
| | - William M Gallagher
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Frankie A Holmes
- Texas Oncology-Memorial Hermann Memorial City, US Oncology Research, Houston, -Texas
| | - Virginia Espina
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Lance Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Bryan T Hennessy
- RCSI Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Education & Research Centre, Beaumont Hospital, Beaumont, Dublin, Ireland.,Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Kenneth J O'Byrne
- Princess Alexandra Hospital, Translational Research Institute and Queensland University of Technology, Brisbane, Queensland, Australia
| | - Max Hasmann
- Roche Innovation Center Penzberg, Roche Diagnostics GmbH, Penzberg, Germany
| | | | - Norma O'Donovan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland
| | - John Crown
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Leinster, Ireland.,Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
4
|
Bie L, Luo S, Li D, Wei Y, Mu Y, Chen X, Wang S, Guo P, Lu X. HOTAIR Competitively Binds MiRNA330 as a Molecular Sponge to Increase the Resistance of Gastric Cancer to Trastuzumab. Curr Cancer Drug Targets 2020; 20:700-709. [PMID: 32364078 DOI: 10.2174/1568009620666200504114000] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND HOTAIR, one of the most widely studied long non-coding RNAs in tumors, is closely related to tumor proliferation, migration, invasion and chemoresistance. OBJECTIVE Here, we studied the mechanism behind proliferation and chemoresistance processes. METHODS A total of 75 samples were collected from patients who underwent surgical resection of their gastric cancer and received trastuzumab treatment. Primary cells were isolated and cultured. We also developed a cell line overexpressing HOTAIR by constructing a lentiviral vector. These cell lines were studied using an array of established biomolecular methods. RESULTS We found that HOTAIR levels were inversely associated with sensitivity to trastuzumab in gastric cancer and that overexpression of HOTAIR can promote the proliferation and invasion of gastric cancer cells. The sensitivity of cells overexpressing HOTAIR to two different types of human epidermal growth factor receptor 2 (HER2) inhibitors (trastuzumab and afatinib) showed that overexpression of HOTAIR is specific for trastuzumab resistance. Furthermore, luciferase reporter gene assay and western blot assay showed that there is a HOTAIR-miRNA330-ERBB4 competitive endogenous RNA regulatory network with miRNA330 as the core. CONCLUSION HOTAIR can not only promote tumor proliferation but also enhance the resistance of tumor cells to drugs. Our experimental data not only showed strong expression of HOTAIR in gastric cancer, but also that strong expression of HOTAIR caused the sensitivity of gastric cancer cells to trastuzumab, which is a useful reference for postoperative medication.
Collapse
Affiliation(s)
- Liangyu Bie
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, Henan Province, China
| | - Suxia Luo
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, Henan Province, China
| | - Dan Li
- Department of General Surgery, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, Henan Province, China
| | - Yan Wei
- Department of Pathology, Nanyang Medical College, Nanyang, Henan province, China
| | - Yu Mu
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, Henan Province, China
| | - Xiaobing Chen
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, Henan Province, China
| | - Saiqi Wang
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, Henan Province, China
| | - Ping Guo
- Department of Oncology, The First Affiliated Hospital of Nanyang Medical College, Nanyang, Henan province, China
| | - Xiaoyu Lu
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, Henan province, China
| |
Collapse
|
5
|
Solassol I, Pinguet F, Quantin X. FDA- and EMA-Approved Tyrosine Kinase Inhibitors in Advanced EGFR-Mutated Non-Small Cell Lung Cancer: Safety, Tolerability, Plasma Concentration Monitoring, and Management. Biomolecules 2019; 9:biom9110668. [PMID: 31671561 PMCID: PMC6921037 DOI: 10.3390/biom9110668] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/23/2019] [Accepted: 10/25/2019] [Indexed: 12/31/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the most common form of primary lung cancer. The discovery of several oncogenic driver mutations in patients with NSCLC has allowed the development of personalized treatments based on these specific molecular alterations, in particular in the tyrosine kinase (TK) domain of the epidermal growth factor receptor (EGFR) gene. Gefitinib, erlotinib, afatinib, and osimertinib are TK inhibitors (TKIs) that specifically target EGFR and are currently approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) as first line treatment for sensitive EGFR-mutant patients. However, these four drugs are associated with severe adverse events (AEs) that can significantly impact patient health-related quality of life and patient monitoring. EGFR-TKIs are commonly used together with other types of medication that can substantially interact. Here, we review approaches used for the management of TKI-AEs in patients with advanced NSCLC to promote the benefits of treatments and minimize the risk of TKI treatment discontinuation. We also consider potential TKI–drug interactions and discuss the usefulness of plasma concentration monitoring TKIs based on chromatographic and mass spectrometry approaches to guide clinical decision-making. Adjusting the most appropriate therapeutic strategies and drug doses may improve the performance therapy and prognosis of patients with advanced EGFR-mutated NSCLC.
Collapse
Affiliation(s)
- Isabelle Solassol
- Unité de Recherche Translationnelle, Institut du Cancer de Montpellier (ICM), 34000 Montpellier, France.
- Département de Pharmacie, Institut du Cancer de Montpellier (ICM), 34000 Montpellier, France.
| | - Frédéric Pinguet
- Département de Pharmacie, Institut du Cancer de Montpellier (ICM), 34000 Montpellier, France.
| | - Xavier Quantin
- Service d'Oncologie Médicale, Institut du Cancer de Montpellier (ICM), IRCM, INSERM, Univ. Montpellier, 34000 Montpellier, France.
| |
Collapse
|
6
|
Di J, Zheng B, Kong Q, Jiang Y, Liu S, Yang Y, Han X, Sheng Y, Zhang Y, Cheng L, Han J. Prioritization of candidate cancer drugs based on a drug functional similarity network constructed by integrating pathway activities and drug activities. Mol Oncol 2019; 13:2259-2277. [PMID: 31408580 PMCID: PMC6763777 DOI: 10.1002/1878-0261.12564] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/13/2019] [Accepted: 08/12/2019] [Indexed: 12/27/2022] Open
Abstract
Due to the speed, efficiency, relative risk, and lower costs compared to traditional drug discovery, the prioritization of candidate drugs for repurposing against cancers of interest has attracted the attention of experts in recent years. Herein, we present a powerful computational approach, termed prioritization of candidate drugs (PriorCD), for the prioritization of candidate cancer drugs based on a global network propagation algorithm and a drug–drug functional similarity network constructed by integrating pathway activity profiles and drug activity profiles. This provides a new approach to drug repurposing by first considering the drug functional similarities at the pathway level. The performance of PriorCD in drug repurposing was evaluated by using drug datasets of breast cancer and ovarian cancer. Cross‐validation tests on the drugs approved for the treatment of these cancers indicated that our approach can achieve area under receiver‐operating characteristic curve (AUROC) values greater than 0.82. Furthermore, literature searches validated our results, and comparison with other classical gene‐based repurposing methods indicated that our pathway‐level PriorCD is comparatively more effective at prioritizing candidate drugs with similar therapeutic effects. We hope that our study will be of benefit to the field of drug discovery. In order to expand the usage of PriorCD, a freely available R‐based package, PriorCD, has been developed to prioritize candidate anticancer drugs for drug repurposing.
Collapse
Affiliation(s)
- Jieyi Di
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Baotong Zheng
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Qingfei Kong
- Department of Neurobiology, Harbin Medical University, China
| | - Ying Jiang
- College of Basic Medical Science, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siyao Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Yang Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Xudong Han
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Yuqi Sheng
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| |
Collapse
|
7
|
Jeibouei S, Akbari ME, Kalbasi A, Aref AR, Ajoudanian M, Rezvani A, Zali H. Personalized medicine in breast cancer: pharmacogenomics approaches. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2019; 12:59-73. [PMID: 31213877 PMCID: PMC6549747 DOI: 10.2147/pgpm.s167886] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/27/2019] [Indexed: 12/14/2022]
Abstract
Abstract: Breast cancer is the fifth cause of cancer death among women worldwide and represents a global health concern due to the lack of effective therapeutic regimens that could be applied to all disease groups. Nowadays, strategies based on pharmacogenomics constitute novel approaches that minimize toxicity while maximizing drug efficacy; this being of high importance in the oncology setting. Besides, genetic profiling of malignant tumors can lead to the development of targeted therapies to be included in effective drug regimens. Advances in molecular diagnostics have revealed that breast cancer is a multifaceted disease, characterized by inter-tumoral and intra-tumoral heterogeneity and, unlike the past, molecular classifications based on the expression of individual biomarkers have led to devising novel therapeutic strategies that improve patient survival. In this review, we report and discuss the molecular classification of breast cancer subtypes, the heterogeneity resource, and the advantages and disadvantages of current drug regimens with consideration of pharmacogenomics in response and resistance to treatment.
Collapse
Affiliation(s)
- Shabnam Jeibouei
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Alireza Kalbasi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mohammad Ajoudanian
- Department of Tissue Engineering and Applied Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Rezvani
- Department of Hematology, Medical Oncology and Stem Cell Transplantation, Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hakimeh Zali
- Proteomics Research Centre, Department of Tissue Engineering and Applied Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Abstract
Afatinib is an oral, irreversible ErbB family blocker that covalently binds to the kinase domains of epidermal growth factor receptor (EGFR), human EGFRs (HER) 2, and HER4, resulting in irreversible inhibition of tyrosine kinase autophosphorylation. Studies in healthy volunteers and patients with advanced solid tumours have shown that once-daily afatinib has time-independent pharmacokinetic characteristics. Maximum plasma concentrations of afatinib are reached approximately 2–5 h after oral administration and thereafter decline, at least bi-exponentially. Food reduces total exposure to afatinib. Over the clinical dose range of 20–50 mg, afatinib exposure increases slightly more than dose proportional. Afatinib metabolism is minimal, with unchanged drug predominantly excreted in the faeces and approximately 5 % in urine. Apart from the parent drug afatinib, the major circulation species in human plasma are the covalently bound adducts to plasma protein. The effective elimination half-life is approximately 37 h, consistent with an accumulation of drug exposure by 2.5- to 3.4-fold based on area under the plasma concentration–time curve (AUC) after multiple dosing. The pharmacokinetic profile of afatinib is consistent across a range of patient populations. Age, ethnicity, smoking status and hepatic function had no influence on afatinib pharmacokinetics, while females and patients with low body weight had increased exposure to afatinib. Renal function is correlated with afatinib exposure, but, as for sex and body weight, the effect size for patients with severe renal impairment (approximately 50 % increase in AUC) is only mildly relative to the extent of unexplained interpatient variability in afatinib exposure. Afatinib has a low potential as a victim or perpetrator of drug–drug interactions, especially with cytochrome P450-modulating agents. However, concomitant treatment with potent inhibitors or inducers of the P-glycoprotein transporter can affect the pharmacokinetics of afatinib. At a dose of 50 mg, afatinib does not have proarrhythmic potential.
Collapse
|
9
|
Martel S, Maurer C, Lambertini M, Pondé N, De Azambuja E. Breast cancer treatment-induced cardiotoxicity. Expert Opin Drug Saf 2017; 16:1021-1038. [PMID: 28697311 DOI: 10.1080/14740338.2017.1351541] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Breast cancer is the most frequent cancer affecting women worldwide. In every setting, the majority of women are treated with an evergrowing arsenal of therapeutic agents that have greatly improved their outcomes. However, these therapies can also be associated with significant adverse events. Areas covered: This review aims to thoroughly describe the current state of the evidence regarding the potential cardiotoxicity of agents commonly used in the treatment of breast cancer. These include chemotherapeutic agents, anti-HER2 therapies and CDK4/6 and mTOR inhibitors. Furthermore, issues related to the risk stratification and monitoring tools are explored. Expert opinion: Anthracycline- and trastuzumab-related cardiac toxicities have been extensively studied. Substantial evidence is now available concerning additional anti-HER2 agents such as pertuzumab, T-DM1 and tyrosine kinase inhibitors; overall, the cardiotoxicity profile is reassuring. Cardiac events due to endocrine therapy are mostly ischemic and, in the context of prolonged therapy, need specific attention. Novel agents implicated in the treatment of hormone receptor-positive disease are potentially arrhythmogenic and the exact risk will need to be further refined. As for today, assessment of baseline risk factors prior to treatment initiation and cardiac imaging before and during treatment remains the optimal way to prevent cardiac dysfunction. Cardioprotective therapy in primary prevention is still a matter of debate.
Collapse
Affiliation(s)
- Samuel Martel
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium.,b Département d'hémato-oncologie , CISSS Montérégie centre/Hôpital Charles Lemoyne, centre affilié de l'Université de Sherbrooke , Greenfield Park , Qc , Canada
| | - Christian Maurer
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium.,c Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn , University of Cologne , Cologne , Germany
| | - Matteo Lambertini
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium.,d Breast Cancer Translational Research Laboratory, Institut Jules Bordet , Université Libre de Bruxelles (U.L.B) , Brussels , Belgium
| | - Noam Pondé
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium
| | - Evandro De Azambuja
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium
| |
Collapse
|
10
|
Localisation Microscopy of Breast Epithelial ErbB-2 Receptors and Gap Junctions: Trafficking after γ-Irradiation, Neuregulin-1β, and Trastuzumab Application. Int J Mol Sci 2017; 18:ijms18020362. [PMID: 28208769 PMCID: PMC5343897 DOI: 10.3390/ijms18020362] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/28/2017] [Accepted: 01/30/2017] [Indexed: 12/28/2022] Open
Abstract
In cancer, vulnerable breast epithelium malignance tendency correlates with number and activation of ErbB receptor tyrosine kinases. In the presented work, we observe ErbB receptors activated by irradiation-induced DNA injury or neuregulin-1β application, or alternatively, attenuated by a therapeutic antibody using high resolution fluorescence localization microscopy. The gap junction turnover coinciding with ErbB receptor activation and co-transport is simultaneously recorded. DNA injury caused by 4 Gray of 6 MeV photon γ-irradiation or alternatively neuregulin-1β application mobilized ErbB receptors in a nucleograde fashion—a process attenuated by trastuzumab antibody application. This was accompanied by increased receptor density, indicating packing into transport units. Factors mobilizing ErbB receptors also mobilized plasma membrane resident gap junction channels. The time course of ErbB receptor activation and gap junction mobilization recapitulates the time course of non-homologous end-joining DNA repair. We explain our findings under terms of DNA injury-induced membrane receptor tyrosine kinase activation and retrograde trafficking. In addition, we interpret the phenomenon of retrograde co-trafficking of gap junction connexons stimulated by ErbB receptor activation.
Collapse
|
11
|
Abstract
Background The drug discovery and development pipeline is a long and arduous process that inevitably hampers rapid drug development. Therefore, strategies to improve the efficiency of drug development are urgently needed to enable effective drugs to enter the clinic. Precision medicine has demonstrated that genetic features of cancer cells can be used for predicting drug response, and emerging evidence suggest that gene-drug connections could be predicted more accurately by exploring the cumulative effects of many genes simultaneously. Results We developed DeSigN, a web-based tool for predicting drug efficacy against cancer cell lines using gene expression patterns. The algorithm correlates phenotype-specific gene signatures derived from differentially expressed genes with pre-defined gene expression profiles associated with drug response data (IC50) from 140 drugs. DeSigN successfully predicted the right drug sensitivity outcome in four published GEO studies. Additionally, it predicted bosutinib, a Src/Abl kinase inhibitor, as a sensitive inhibitor for oral squamous cell carcinoma (OSCC) cell lines. In vitro validation of bosutinib in OSCC cell lines demonstrated that indeed, these cell lines were sensitive to bosutinib with IC50 of 0.8–1.2 μM. As further confirmation, we demonstrated experimentally that bosutinib has anti-proliferative activity in OSCC cell lines, demonstrating that DeSigN was able to robustly predict drug that could be beneficial for tumour control. Conclusions DeSigN is a robust method that is useful for the identification of candidate drugs using an input gene signature obtained from gene expression analysis. This user-friendly platform could be used to identify drugs with unanticipated efficacy against cancer cell lines of interest, and therefore could be used for the repurposing of drugs, thus improving the efficiency of drug development. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3260-7) contains supplementary material, which is available to authorized users.
Collapse
|