1
|
Qu F, Lu R, Wu X, Liu Q, Zha M, Li H, Yuan Y, Han Z, Cai D, Huang X, Yin Y, Li W. Efficacy and safety of RC48-ADC in HER2-positive and HER2-low metastatic breast cancer: a multicenter, real-world study. Front Oncol 2024; 14:1435485. [PMID: 39582543 PMCID: PMC11582051 DOI: 10.3389/fonc.2024.1435485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/22/2024] [Indexed: 11/26/2024] Open
Abstract
Background A standard treatment recommendation for third-line and subsequent treatments for advanced HER2-positive breast cancer is still missing, especially for low HER2 expression. Nevertheless, there is evidence that these patients might benefits from antibody-drug conjugates (ADCs) treatment. Therefore, this study aimed to evaluate the clinical efficacy, safety, and factors affecting efficacy of Disitamab Vedotin (RC48) for treating HER2-positive and HER2-low metastatic breast cancer (MBC) in the real-world setting. Methods A retrospective study at five clinical sites was conducted in China, enrolling MBC patients treated with RC48 from July 01, 2021 and May 31, 2023. Patient demographics, treatment patterns, and adverse events (AEs) were recorded and analyzed. Results A total of 154 patients were included: 104 (67.53%) patients with HER2-positive and 50 (32.47%) patients with HER2-low MBC. The median progression-free survival (mPFS) was 5.06 months. The objective response rate (ORR) and disease control rate (DCR) were 36.36% and 68.83%, respectively. HER2-positive patients exhibited a mPFS of 5.93 and an ORR of 41.35%. In contrast, patients with low-HER2 had a mPFS of 4.28 months and an ORR of 26.00%. The most common AEs included neutropenia (54.55%), increased AST (53.25%), leukopenia (51.95%), and fatigue (43.51%), mostly graded mild to moderate (grade 1-2). Conclusions This extensive study in China demonstrated that RC48 has excellent therapeutic potential for both HER2-positive and HER2-low MBC with a favorable safety profile. The study also suggests that combination therapy significantly boosts efficacy beyond monotherapy, indicating a promising avenue for future ADC development.
Collapse
Affiliation(s)
- Fei Qu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Rongrong Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Xinyu Wu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Qian Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Mengyao Zha
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Huihui Li
- Department of Breast Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yuan Yuan
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengxiang Han
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dongyan Cai
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiang Huang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Kang Z, Jin Y, Yu H, Li S, Qi Y. Relative efficacy of antibody-drug conjugates and other anti-HER2 treatments on survival in HER2-positive advanced breast cancer: a systematic review and meta-analysis. BMC Cancer 2024; 24:708. [PMID: 38851684 PMCID: PMC11162572 DOI: 10.1186/s12885-024-12478-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 06/04/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Novel antibody-drug conjugates (ADCs) drugs present a promising anti-cancer treatment, although survival benefits for HER2-positive advanced breast cancer (BC) remain controversial. The aim of this meta-analysis was to evaluate the comparative effect of ADCs and other anti-HER2 therapy on progression-free survival (PFS) and overall survival (OS) for treatment of HER2-positive locally advanced or metastatic BC. METHODS Relevant randomized controlled trials (RCTs) were retrieved from five databases. The risk of bias was assessed with the Cochrane Collaboration's tool for RCTs by RevMan5.4 software. The hazard ratio (HR) and 95% confidence intervals (CIs) were extracted to evaluate the benefit of ADCs on PFS and OS in HER2-positive advanced BC by meta-analysis. RESULTS Meta-analysis of six RCTs with 3870 patients revealed that ADCs significantly improved PFS (HR: 0.63, 95% CI: 0.49-0.80, P = 0.0002) and OS (HR: 0.79, 95% CI: 0.72-0.86, P < 0.0001) of patients with HER2-positive locally advanced or metastatic BC. Subgroup analysis showed that PFS and OS were obviously prolonged for patients who previously received HER2-targeted therapy. Sensitivity analysis and publication bias suggested that the results were stable and reliable. CONCLUSION Statistically significant benefits for PFS and OS were observed with ADCs in HER2-positive locally advanced or metastatic BC, especially for those who received prior anti-HER2 treatment.
Collapse
Affiliation(s)
- Zian Kang
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, 110042, China
| | - Yuqing Jin
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Huihui Yu
- Department of Cancer Prevention and Control, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Su Li
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, 110042, China
| | - Yingjie Qi
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, 110042, China.
| |
Collapse
|
3
|
Nasrazadani A, Marti JLG, Lathrop K, Restrepo A, Leu SY, Bhat G, Brufsky A. Poziotinib treatment in patients with HER2-positive advanced breast cancer who have received prior anti-HER2 regimens. Breast Cancer Res Treat 2024; 205:29-37. [PMID: 38261228 DOI: 10.1007/s10549-023-07236-z] [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: 05/19/2023] [Accepted: 11/22/2023] [Indexed: 01/24/2024]
Abstract
PURPOSE Poziotinib is an irreversible pan-inhibitor of the human epidermal growth factor receptor (HER) that has shown acceptable tolerability and antitumor activity in phase I and II trials in patients with advanced solid tumors. In the present open-label, multicenter phase II study, we demonstrate safety, tolerability, and preliminary efficacy data from two different dosing schedules in patients with HER2-positive advanced breast cancer. PATIENTS AND METHODS Patients who had received at least two prior HER2-directed therapy lines for advanced disease, received 24 mg poziotinib on an intermittent dosing schedule (cohort 1) or 16 mg poziotinib once daily on a continuous dosing schedule (cohort 2). The primary endpoint was overall response rate (ORR). Secondary endpoints were progression-free survival (PFS), disease control rate (DCR), and time to progression (TTP). Secondary endpoints additionally included safety and pharmacokinetics. RESULTS A total of 67 patients were enrolled. The ORR was 30% in both groups (p = 0.98). DCR was 60% vs 78% (p = 0.15) and median PFS and TTP were 4.1 vs 4.9 months (both p = 0.30) for cohorts 1 and 2, respectively. The most common treatment related adverse events (AEs) of any grade included diarrhea (88% vs 85%, p = 0.76), rash (88% vs 88%, p = 0.96), and stomatitis (64% vs 56%, p = 0.52), with grade 3-4 diarrhea occurring in 33% vs 32% of patients (p = 0.93) and grade 3-4 rash in 27% vs 35% of patients (p = 0.48) in cohort 1 vs cohort 2, respectively. CONCLUSION Poziotinib demonstrated evidence of clinical activity in patients with pre-treated HER2-positive advanced breast cancer, although high levels of toxicity may preclude further studies at this time.
Collapse
Affiliation(s)
- Azadeh Nasrazadani
- Division of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Kate Lathrop
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | | | | | - Adam Brufsky
- Magee-Womens Hospital of UPMC, 300 Halket St, Pittsbugh, PA, 15213, USA.
| |
Collapse
|
4
|
Genna A, Alter J, Poletti M, Meirson T, Sneh T, Gendler M, Saleev N, Karagiannis GS, Wang Y, Cox D, Entenberg D, Oktay MH, Korcsmaros T, Condeelis JS, Gil-Henn H. FAK family proteins regulate in vivo breast cancer metastasis via distinct mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.27.564212. [PMID: 37961438 PMCID: PMC10634866 DOI: 10.1101/2023.10.27.564212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Breast cancer is the most commonly diagnosed malignancy and the major leading cause of tumor-related deaths in women. It is estimated that the majority of breast tumor-related deaths are a consequence of metastasis, to which no cure exists at present. The FAK family proteins Proline-rich tyrosine kinase (PYK2) and focal adhesion kinase (FAK) are highly expressed in breast cancer, but the exact cellular and signaling mechanisms by which they regulate in vivo tumor cell invasiveness and consequent metastatic dissemination are mostly unknown. Using a PYK2 and FAK knockdown xenograft model we show here, for the first time, that ablation of either PYK2 or FAK decreases primary tumor size and significantly reduces Tumor MicroEnvironment of Metastasis (TMEM) doorway activation, leading to decreased intravasation and reduced spontaneous lung metastasis. Intravital imaging analysis further demonstrates that PYK2, but not FAK, regulates a motility phenotype switch between focal adhesion-mediated fast motility and invadopodia-dependent, ECM-degradation associated slow motility within the primary tumor. Furthermore, we validate our in vivo and intravital imaging results with integrated transcriptomic and proteomic data analysis from xenograft knockdown tumors and reveal new and distinct pathways by which these two homologous kinases regulate breast tumor cell invasiveness and consequent metastatic dissemination. Our findings identify PYK2 and FAK as novel mediators of mammary tumor progression and metastasis and as candidate therapeutic targets for breast cancer metastasis.
Collapse
|
5
|
Peerzada MN, Hamdy R, Rizvi MA, Verma S. Privileged Scaffolds in Drug Discovery against Human Epidermal Growth Factor Receptor 2 for Cancer Treatment. Curr Pharm Des 2023; 29:3563-3578. [PMID: 38141192 DOI: 10.2174/0113816128283615231218094706] [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: 09/22/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 12/25/2023]
Abstract
HER2 is the membrane receptor tyrosine kinase showing overexpression in several human malignancies, particularly breast cancer. HER2 overexpression causes the activation of Ras- MAPK and PI3K/Akt/ NF-κB cellular signal transduction pathways that lead to cancer development and progression. HER2 is, therefore, presumed as one of the key targets for the development of tumor-specific therapies. Several preclinical have been developed that function by inhibiting the HER2 tyrosine kinase activity through the prevention of the dimerization process. Most HER2 inhibitors act as ATP competitors and prevent the process of phosphorylation, and abort the cell cycle progression and proliferation. In this review, the clinical drug candidates and potent pre-clinical newly developed molecules are described, and the core chemical scaffolds typically responsible for anti-HER2 activity are deciphered. In addition, the monoclonal antibodies that are either used in monotherapy or in combination therapy against HER2-positive cancer are briefly described. The identified key moieties in this study could result in the discovery of more effective HER2-targeted anticancer drug molecules and circumvent the development of resistance by HER2-specific chemotherapeutics in the future.
Collapse
Affiliation(s)
- Mudasir Nabi Peerzada
- Tumor Biology and Drug Discovery Laboratory, National Institute of Pathology, Indian Council of Medical Research, Safdarjang Hospital Campus, New Delhi 110029, India
| | - Rania Hamdy
- Research Institute for Science and Engineering (RISE), University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | | | - Saurabh Verma
- Tumor Biology and Drug Discovery Laboratory, National Institute of Pathology, Indian Council of Medical Research, Safdarjang Hospital Campus, New Delhi 110029, India
| |
Collapse
|
6
|
Johnson KC, Quiroga D, Sudheendra P, Wesolowski R. Treatment of small (T1mic, T1a, and T1b) node-negative HER2+ breast cancer - a review of current evidence for and against the use of anti-HER2 treatment regimens. Expert Rev Anticancer Ther 2022; 22:505-522. [PMID: 35389302 PMCID: PMC9156575 DOI: 10.1080/14737140.2022.2063844] [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: 11/04/2022]
Abstract
INTRODUCTION Since the advent of anti-HER2 therapies, evidence surrounding adjuvant treatment of small (T1mic, T1a, and T1b), node-negative, HER2-positive breast cancer (HER2+BC) has remained limited. Practices vary widely between institutions with little known regarding the added benefit of systemic therapy, including cytotoxic chemotherapy and HER2-directed treatments. Our group has set out to perform an extensive review of available literature on this topic. AREAS COVERED In this review, we examined HER2 biology, anti-HER therapies, outcome definitions, and available prospective and retrospective data surrounding the use of adjuvant therapy in those with small, node-negative, HER2+BC. For outcomes, we primarily explored breast cancer-specific survival (BCSS), invasive disease-free survival (iDFS), and overall survival (OS). We also investigated the incidence of adverse events with a particular focus on symptomatic and asymptomatic declines in left ventricular ejection fraction. EXPERT OPINION Retrospective data will likely be the main driver for future treatment decisions. Given what we know, high risk T1b and T1c subgroups derive measurable added benefit from HER2-guided combination therapies but it's not clear whether these benefits outweigh known risks associated with this combination therapy. For tumors ≤0.5cm (T1mic and T1a), treatment remains highly controversial with limited evidence available through retrospective analysis that suggest over-treatment may be occurring.
Collapse
Affiliation(s)
- Kai Cc Johnson
- Stefanie Spielman Comprehensive Breast Center, Columbus, OH, USA
| | - Dionisia Quiroga
- Stefanie Spielman Comprehensive Breast Center, Columbus, OH, USA.,Pelotonia Institute for Immuno-Oncology, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Robert Wesolowski
- Stefanie Spielman Comprehensive Breast Center, Columbus, OH, USA.,Pelotonia Institute for Immuno-Oncology, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
7
|
Bléry M, Mrabet-Kraiem M, Morel A, Lhospice F, Bregeon D, Bonnafous C, Gauthier L, Rossi B, Remark R, Cornen S, Anceriz N, Viaud N, Trichard S, Carpentier S, Joulin-Giet A, Grondin G, Liptakova V, Kim Y, Daniel L, Haffner A, Macagno N, Pouyet L, Perrot I, Paturel C, Morel Y, Steinle A, Romagné F, Narni-Mancinelli E, Vivier E. Targeting MICA/B with cytotoxic therapeutic antibodies leads to tumor control. OPEN RESEARCH EUROPE 2021; 1:107. [PMID: 35967081 PMCID: PMC7613279 DOI: 10.12688/openreseurope.13314.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/18/2021] [Indexed: 11/20/2022]
Abstract
Background: MICA and MICB are tightly regulated stress-induced proteins that trigger the immune system by binding to the activating receptor NKG2D on cytotoxic lymphocytes. MICA and MICB are highly polymorphic molecules with prevalent expression on several types of solid tumors and limited expression in normal/healthy tissues, making them attractive targets for therapeutic intervention. Methods: We have generated a series of anti-MICA and MICB cross-reactive antibodies with the unique feature of binding to the most prevalent isoforms of both these molecules. Results: The anti-MICA and MICB antibody MICAB1, a human IgG1 Fc-engineered monoclonal antibody (mAb), displayed potent antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) of MICA/B-expressing tumor cells in vitro. However, it showed insufficient efficiency against solid tumors in vivo, which prompted the development of antibody-drug conjugates (ADC). Indeed, optimal tumor control was achieved with MICAB1-ADC format in several solid tumor models, including patient-derived xenografts (PDX) and carcinogen-induced tumors in immunocompetent MICAgen transgenic mice. Conclusions: These data indicate that MICA and MICB are promising targets for cytotoxic immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Veronika Liptakova
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Younghoon Kim
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Laurent Daniel
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Marseille, France
| | - Aurélie Haffner
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Marseille, France
| | - Nicolas Macagno
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Marseille, France
| | | | | | | | | | - Alexander Steinle
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Frankfurt am Main, Germany
| | | | | | - Eric Vivier
- Innate Pharma, Marseille, France
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Marseille, France
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| |
Collapse
|
8
|
Bléry M, Mrabet-Kraiem M, Morel A, Lhospice F, Bregeon D, Bonnafous C, Gauthier L, Rossi B, Remark R, Cornen S, Anceriz N, Viaud N, Trichard S, Carpentier S, Joulin-Giet A, Grondin G, Liptakova V, Kim Y, Daniel L, Haffner A, Macagno N, Pouyet L, Perrot I, Paturel C, Morel Y, Steinle A, Romagné F, Narni-Mancinelli E, Vivier E. Targeting MICA/B with cytotoxic therapeutic antibodies leads to tumor control. OPEN RESEARCH EUROPE 2021; 1:107. [PMID: 35967081 PMCID: PMC7613279 DOI: 10.12688/openreseurope.13314.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/18/2021] [Indexed: 02/16/2024]
Abstract
Background: MICA and MICB are tightly regulated stress-induced proteins that trigger the immune system by binding to the activating receptor NKG2D on cytotoxic lymphocytes. MICA and MICB are highly polymorphic molecules with prevalent expression on several types of solid tumors and limited expression in normal/healthy tissues, making them attractive targets for therapeutic intervention. Methods: We have generated a series of anti-MICA and MICB cross-reactive antibodies with the unique feature of binding to the most prevalent isoforms of both these molecules. Results: The anti-MICA and MICB antibody MICAB1, a human IgG1 Fc-engineered monoclonal antibody (mAb), displayed potent antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) of MICA/B-expressing tumor cells in vitro. However, it showed insufficient efficiency against solid tumors in vivo, which prompted the development of antibody-drug conjugates (ADC). Indeed, optimal tumor control was achieved with MICAB1-ADC format in several solid tumor models, including patient-derived xenografts (PDX) and carcinogen-induced tumors in immunocompetent MICAgen transgenic mice. Conclusions: These data indicate that MICA and MICB are promising targets for cytotoxic immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Veronika Liptakova
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Younghoon Kim
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Laurent Daniel
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Marseille, France
| | - Aurélie Haffner
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Marseille, France
| | - Nicolas Macagno
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Marseille, France
| | | | | | | | | | - Alexander Steinle
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Frankfurt am Main, Germany
| | | | | | - Eric Vivier
- Innate Pharma, Marseille, France
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Marseille, France
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| |
Collapse
|
9
|
Guardia C, Bianchini G, Arpí-LLucià O, Menendez S, Casadevall D, Galbardi B, Dugo M, Servitja S, Montero JC, Soria-Jiménez L, Sabbaghi M, Peña R, Madoz-Gúrpide J, Lloveras B, Lluch A, Eroles P, Arribas J, Pandiella A, Gianni L, Rojo F, Rovira A, Albanell J. Preclinical and Clinical Characterization of Fibroblast-derived Neuregulin-1 on Trastuzumab and Pertuzumab Activity in HER2-positive Breast Cancer. Clin Cancer Res 2021; 27:5096-5108. [PMID: 34385295 DOI: 10.1158/1078-0432.ccr-20-2915] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 05/29/2021] [Accepted: 07/12/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE To characterize expression of neuregulin-1 (NRG1), an HER3 ligand, in HER2-positive breast cancer and its relation with the efficacy of trastuzumab with or without pertuzumab. EXPERIMENTAL DESIGN Characterization of NRG1 expression in tumor cell lines, in tumor specimens, and in cancer-associated fibroblasts (CAFs). Patient-derived CAFs were used to investigate NRG1 impact on the activity of trastuzumab with or without pertuzumab in HER2-positive breast cancer cells. The relationship between NRG1 expression and pathologic response to anti-HER2-based neoadjuvant therapy was assessed in a retrospective patient cohort and in the NeoSphere trial. RESULTS NRG1 was expressed in HER2-positive breast cancer-derived fibroblasts at significantly higher levels than in cancer cells. NRG1 and the conditioned media (CM) from CAFs phosphorylated HER3 and AKT in cancer cells and mediated trastuzumab resistance. Stable genetic depletion of NRG1 from CAFs overcame trastuzumab resistance. Pertuzumab effectively suppressed trastuzumab resistance mediated by either NRG1 or CAF's CM. NRG1 engaged an epithelial-to-mesenchymal transition that was prevented by trastuzumab and pertuzumab. In clinical samples, stromal and/or tumor cell expression of NRG1 determined by immunohistochemistry was uncommon (13.2%) yet significantly linked with residual disease following trastuzumab-based neoadjuvant therapy. In the NeoSphere trial, the magnitude of the difference of pathologic complete response rates favoring the pertuzumab arm was higher in the NRG1-high group. CONCLUSIONS CAF-derived NRG1 mediates trastuzumab resistance through HER3/AKT, which might be reverted by pertuzumab. In patients with HER2-positive breast cancer, high expression of NRG1 was associated to poor response to trastuzumab, but not in combination with pertuzumab.
Collapse
Affiliation(s)
- Cristina Guardia
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | | | - Oriol Arpí-LLucià
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Silvia Menendez
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - David Casadevall
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Barbara Galbardi
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Matteo Dugo
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Sonia Servitja
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | | | - Luis Soria-Jiménez
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - MohammadA Sabbaghi
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Raul Peña
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Juan Madoz-Gúrpide
- Department of Pathology, IIS-Fundación Jiménez Díaz-CIBERONC, Madrid, Spain
| | - Belen Lloveras
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Ana Lluch
- INCLIVA Biomedical Research Institute, Hospital Clínico de Valencia-CIBERONC, Universitat de València, Barcelona, Spain
| | - Pilar Eroles
- INCLIVA Biomedical Research Institute, Hospital Clínico de Valencia-CIBERONC, Universitat de València, Barcelona, Spain.,INCLIVA, Biomedical Research Institute, Valencia, Spain
| | - Joaquin Arribas
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer, IBSAL-CSIC-CIBERONC, Salamanca, Spain
| | | | - Federico Rojo
- Department of Pathology, IIS-Fundación Jiménez Díaz-CIBERONC, Madrid, Spain
| | - Ana Rovira
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain. .,Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
10
|
Spring LM, Clark SL, Li T, Goel S, Tayob N, Viscosi E, Abraham E, Juric D, Isakoff SJ, Mayer E, Moy B, Supko JG, Tolaney SM, Bardia A. Phase 1b clinical trial of ado-trastuzumab emtansine and ribociclib for HER2-positive metastatic breast cancer. NPJ Breast Cancer 2021; 7:103. [PMID: 34349115 PMCID: PMC8339067 DOI: 10.1038/s41523-021-00311-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/25/2021] [Indexed: 11/18/2022] Open
Abstract
Patients with HER2+ metastatic breast cancer are often treated with a multitude of therapies in the metastatic setting, and additional strategies to prolong responses to anti-HER2 therapies are needed. Preclinical evidence suggests synergy between cyclin-dependent kinase 4 and 6 (CDK 4/6) inhibitors and anti-HER2 therapies. We conducted a phase 1b study of ribociclib and ado-trastuzumab emtansine (T-DM1) in patients with advanced/metastatic HER2-positive breast cancer previously treated with trastuzumab and a taxane in any setting, with four or fewer prior lines of therapy in the metastatic setting. A standard 3 + 3 dose-escalation design was used to evaluate various doses of ribociclib in combination with T-DM1, starting at 300 mg. The primary objective was to determine the maximum tolerated dose and/or recommended phase 2 dose (RP2D) of ribociclib in combination with T-DM1. A total of 12 patients were enrolled. During dose-escalation, patients received doses of ribociclib of 300 mg (n = 3), 400 mg (n = 3), 500 mg (n = 3), and 600 mg (n = 3). No dose-limiting toxicities were observed. The majority of toxicities were Grade 1 and 2, and the most common Grade 3 toxicities were neutropenia (33%), leukopenia (33%), and anemia (25%). After a median follow-up of 12.4 months, the median PFS was 10.4 months (95% confidence interval, 2.7-19.3). Based on the pharmacokinetic analysis, adverse events, and dose reductions, 400 mg was determined to be the RP2D for ribociclib given on days 8-21 of a 21-day cycle with T-DM1.
Collapse
Affiliation(s)
- Laura M Spring
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Shealagh L Clark
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Tianyu Li
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Shom Goel
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nabihah Tayob
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Elene Viscosi
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Elizabeth Abraham
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Dejan Juric
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Steven J Isakoff
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Erica Mayer
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Beverly Moy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Jeffrey G Supko
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Sara M Tolaney
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
HER2-PI9 and HER2-I12: two novel and functionally active splice variants of the oncogene HER2 in breast cancer. J Cancer Res Clin Oncol 2021; 147:2893-2912. [PMID: 34136934 PMCID: PMC8397700 DOI: 10.1007/s00432-021-03689-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/05/2021] [Indexed: 11/03/2022]
Abstract
In this study, two novel alternative splice variants of HER2, named HER2-PI9 and HER2-I12, were identified in breast cancer cell lines and breast tumour tissues. Whilst HER2-P19 arises from the inclusion of an 117 bp cassette-exon of intron 9 of HER2, HER2-I12 results from intron 12 inclusion. In silico analyses were performed to predict the amino acid sequences of these two HER2 novel variants. To confirm their protein expression, plasmid vectors were generated and transfected into the HER2 negative breast cancer cell line, MCF-7. Additionally, their functional properties in oncogenic signalling were confirmed. Expression of HER2-PI9 and HER2-I12 was successful and matched the in silico predictions. Importantly, these splice variants can modulate the phosphorylation levels of extracellular signal-related kinase 1/2 (ERK1/2) and Akt/protein kinase B (Akt) signalling in MCF-7 breast cancer cells. Enhanced cellular proliferation, migration and invasion were observed in the case of the HER2-I12 expressing model. In human tissues and breast carcinoma tumours both variants were present. This study reveals two novel splice variants of HER2. Additionally, the potential biological activity for HER2-PI9 and HER2-I12 in breast cancer cells is also reported..
Collapse
|
12
|
Janiszewska M, Stein S, Metzger Filho O, Eng J, Kingston NL, Harper NW, Rye IH, Alečković M, Trinh A, Murphy KC, Marangoni E, Cristea S, Oakes B, Winer EP, Krop IE, Russnes HG, Spellman PT, Bucher E, Hu Z, Chin K, Gray JW, Michor F, Polyak K. The impact of tumor epithelial and microenvironmental heterogeneity on treatment responses in HER2+ breast cancer. JCI Insight 2021; 6:147617. [PMID: 33886505 PMCID: PMC8262355 DOI: 10.1172/jci.insight.147617] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/21/2021] [Indexed: 12/20/2022] Open
Abstract
Despite the availability of multiple human epidermal growth factor receptor 2-targeted (HER2-targeted) treatments, therapeutic resistance in HER2+ breast cancer remains a clinical challenge. Intratumor heterogeneity for HER2 and resistance-conferring mutations in the PIK3CA gene (encoding PI3K catalytic subunit α) have been investigated in response and resistance to HER2-targeting agents, while the role of divergent cellular phenotypes and tumor epithelial-stromal cell interactions is less well understood. Here, we assessed the effect of intratumor cellular genetic heterogeneity for ERBB2 (encoding HER2) copy number and PIK3CA mutation on different types of neoadjuvant HER2-targeting therapies and clinical outcome in HER2+ breast cancer. We found that the frequency of cells lacking HER2 was a better predictor of response to HER2-targeted treatment than intratumor heterogeneity. We also compared the efficacy of different therapies in the same tumor using patient-derived xenograft models of heterogeneous HER2+ breast cancer and single-cell approaches. Stromal determinants were better predictors of response than tumor epithelial cells, and we identified alveolar epithelial and fibroblastic reticular cells as well as lymphatic vessel endothelial hyaluronan receptor 1-positive (Lyve1+) macrophages as putative drivers of therapeutic resistance. Our results demonstrate that both preexisting and acquired resistance to HER2-targeting agents involve multiple mechanisms including the tumor microenvironment. Furthermore, our data suggest that intratumor heterogeneity for HER2 should be incorporated into treatment design.
Collapse
Affiliation(s)
- Michalina Janiszewska
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.,Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida, USA
| | - Shayna Stein
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Otto Metzger Filho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer Eng
- OHSU Center for Spatial Systems Biomedicine, Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA.,OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Natalie L Kingston
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Nicholas W Harper
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Inga H Rye
- Department of Pathology, Division of Laboratory Medicine, and Department of Cancer Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Maša Alečković
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Anne Trinh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Katherine C Murphy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Simona Cristea
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Benjamin Oakes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Eric P Winer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Ian E Krop
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Hege G Russnes
- Department of Pathology, Division of Laboratory Medicine, and Department of Cancer Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Paul T Spellman
- OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA.,Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Elmar Bucher
- OHSU Center for Spatial Systems Biomedicine, Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA.,OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Zhi Hu
- OHSU Center for Spatial Systems Biomedicine, Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA.,OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Koei Chin
- OHSU Center for Spatial Systems Biomedicine, Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA.,OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Joe W Gray
- OHSU Center for Spatial Systems Biomedicine, Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA.,OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA.,Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Ludwig Center at Harvard Medical School, Boston, Massachusetts, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.,Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Ludwig Center at Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Wu Y, Liu H, Gong Y, Zhang B, Chen W. ANKRD22 enhances breast cancer cell malignancy by activating the Wnt/β-catenin pathway via modulating NuSAP1 expression. Bosn J Basic Med Sci 2021; 21:294-304. [PMID: 32651974 PMCID: PMC8112564 DOI: 10.17305/bjbms.2020.4701] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is one of the most prevalent malignancies in women worldwide. Although great advancements have been achieved in the diagnosis and treatment of breast cancer, the prognosis of patients with breast cancer is still poor due to distal recurrence and metastasis after surgery. This study aimed to assess the role of ankyrin repeat domain 22 (ANKRD22) in the progression of breast cancer and investigate the molecular mechanism. Using immunohistochemistry, we demonstrated that the expression level of ANKRD22 in human breast cancer tissues was significantly higher than that in normal breast tissues. ANKRD22 knockdown inhibited the proliferation, invasion, and epithelial-mesenchymal transition (EMT) of breast cancer cells, as confirmed by BrdU, colony formation, transwell, and immunoblot assays. Immunoblot assays further indicated that ANKRD22 regulated the expression of nucleolar and spindle-associated protein 1 (NuSAP1) and then caused the activation of Wnt/β-catenin signaling pathway. Moreover, overexpression of NUSAP1 reversed the inhibitory effects of ANKRD22 knockdown on the proliferation, invasion, and EMT of breast cancer cells. In summary, this study demonstrated that ANKRD22 enhanced breast cancer cell malignancy by activating the Wnt/β-catenin pathway via modulating NuSAP1 expression, which might shed light on new therapeutic approaches for breast cancer.
Collapse
Affiliation(s)
- Yange Wu
- Department of Pathology, Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, China
| | - Hongxia Liu
- Department of Pathology, Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, China
| | - Yufeng Gong
- Department of Pathology, Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, China
| | - Bo Zhang
- Department of Pathology, Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, China
| | - Wenxiu Chen
- Department of Pathology, Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, China
| |
Collapse
|
14
|
Jiang H, Li H. Prognostic values of tumoral MMP2 and MMP9 overexpression in breast cancer: a systematic review and meta-analysis. BMC Cancer 2021; 21:149. [PMID: 33568081 PMCID: PMC7877076 DOI: 10.1186/s12885-021-07860-2] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 01/31/2021] [Indexed: 12/13/2022] Open
Abstract
Background Breast cancer (BC) is a leading cause of cancer-related death in females worldwide. Previous studies have demonstrated that matrix metalloproteinases (MMPs) play key roles in metastasis and are associated with survival in various cancers. The prognostic values of MMP2 and MMP9 expression in BC have been investigated, but the results remain controversial. Thus, we performed the present meta-analysis to investigate the associations between MMP2/9 expressions in tumor cells with clinicopathologic features and survival outcome in BC patients. Methods Eligible studies were searched in PubMed, Web of Science, EMBASE, CNKI and Wanfang databases. The associations of MMP2/9 overexpression in tumor cells with overall survival (OS), disease-free survival (DFS) and recurrence-free survival (RFS) were assessed by hazard ratio (HR) and 95% confidence interval (CI). The associations of MMP2/9 overexpression with clinicopathological features were investigated by calculating odds ratio (OR) and 95% CI. Subgroup analysis, sensitivity analysis, meta-regression, and analysis for publication bias were performed. Results A total of 41 studies comprising 6517 patients with primary BC were finally included. MMP2 overexpression was associated with an unfavorable OS (HR = 1.60, 95% CI 1.33 –1.94, P < 0.001) while MMP9 overexpression predicted a shorter OS (HR = 1.52, 95% CI 1.30 –1.77, P < 0.001). MMP2 overexpression conferred a higher risk to distant metastasis (OR = 2.69, 95% CI 1.35–5.39, P = 0.005) and MMP9 overexpression correlated with lymph node metastasis (OR = 2.90, 95% CI 1.86 – 4.53, P < 0.001). Moreover, MMP2 and MMP9 overexpression were both associated with higher clinical stage and histological grade in BC patients. MMP9 overexpression was more frequent in patients with larger tumor sizes. Conclusions Tumoral MMP2 and MMP9 are promising markers for predicting the prognosis in patients with BC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07860-2.
Collapse
Affiliation(s)
- Hanfang Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, No. 52nd Fucheng Road, Haidian District, Beijing, 100142, China
| | - Huiping Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, No. 52nd Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
15
|
Rigiracciolo DC, Cirillo F, Talia M, Muglia L, Gutkind JS, Maggiolini M, Lappano R. Focal Adhesion Kinase Fine Tunes Multifaced Signals toward Breast Cancer Progression. Cancers (Basel) 2021; 13:645. [PMID: 33562737 PMCID: PMC7915897 DOI: 10.3390/cancers13040645] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer represents the most common diagnosed malignancy and the main leading cause of tumor-related death among women worldwide. Therefore, several efforts have been made in order to identify valuable molecular biomarkers for the prognosis and prediction of therapeutic responses in breast tumor patients. In this context, emerging discoveries have indicated that focal adhesion kinase (FAK), a non-receptor tyrosine kinase, might represent a promising target involved in breast tumorigenesis. Of note, high FAK expression and activity have been tightly correlated with a poor clinical outcome and metastatic features in several tumors, including breast cancer. Recently, a role for the integrin-FAK signaling in mechanotransduction has been suggested and the function of FAK within the breast tumor microenvironment has been ascertained toward tumor angiogenesis and vascular permeability. FAK has been also involved in cancer stem cells (CSCs)-mediated initiation, maintenance and therapeutic responses of breast tumors. In addition, the potential of FAK to elicit breast tumor-promoting effects has been even associated with the capability to modulate immune responses. On the basis of these findings, several agents targeting FAK have been exploited in diverse preclinical tumor models. Here, we recapitulate the multifaceted action exerted by FAK and its prognostic significance in breast cancer. Moreover, we highlight the recent clinical evidence regarding the usefulness of FAK inhibitors in the treatment of breast tumors.
Collapse
Affiliation(s)
- Damiano Cosimo Rigiracciolo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| | - Marianna Talia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| | - Lucia Muglia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| | - Jorge Silvio Gutkind
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA;
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| |
Collapse
|
16
|
Hart V, Gautrey H, Kirby J, Tyson-Capper A. HER2 splice variants in breast cancer: investigating their impact on diagnosis and treatment outcomes. Oncotarget 2020; 11:4338-4357. [PMID: 33245725 PMCID: PMC7679030 DOI: 10.18632/oncotarget.27789] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/10/2020] [Indexed: 02/07/2023] Open
Abstract
Overexpression of the HER2 receptor occurs in approximately 20% of breast cancer patients. HER2 positivity is associated with poor prognosis and aggressive tumour phenotypes, which led to rapid progress in HER2 targeted therapeutics and diagnostic testing. Whilst these advances have greatly increased patients' chances of survival, resistance to HER2 targeted therapies, be that intrinsic or acquired, remains a problem. Different forms of the HER2 protein exist within tumours in tandem and can display altered biological activities. Interest in HER2 variants in breast cancer increased when links between resistance to anti-HER2 therapies and a particular variant, Δ16-HER2, were identified. Moreover, the P100 variant potentially reduces the efficacy of the anti-HER2 therapy trastuzumab. Another variant, Herstatin, exhibits 'auto-inhibitory' behaviour. More recently, new HER2 variants have been identified and are currently being assessed for their pro- and anti-cancer properties. It is important when directing the care of patients to consider HER2 variants collectively. This review considers HER2 variants in the context of the tumour environment where multiple variants are co-expressed at altered ratios. This study also provides an up to date account of the landscape of HER2 variants and links this to patterns of resistance against HER2 therapies and treatment plans.
Collapse
Affiliation(s)
- Vic Hart
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Hannah Gautrey
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - John Kirby
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Alison Tyson-Capper
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|