1
|
Xie X, Zhang J, Sun L, Xu S, Ma SS, Wang H, Li X, Xiang Q, Cui L, Liang X. Ultrasound-triggered topical oxygen delivery enhances synergistic sonodynamic and antibody therapies against hypoxic gastric cancer. J Control Release 2025; 380:736-750. [PMID: 39947405 DOI: 10.1016/j.jconrel.2025.02.019] [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: 12/12/2024] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/21/2025]
Abstract
Hypoxia is a common feature of malignant tumors, which can accelerate tumor growth and reduce the sensitivity of chemotherapy and sonodynamic therapy by activating the hypoxia-inducible factor (HIF) signaling pathway. In HER2-positive gastric cancer, HER2 overexpression enhances HIF-1α synthesis, exacerbating hypoxia and impairing sonodynamic therapy. It also reduces trastuzumab-mediated antibody-dependent cytotoxicity, significantly compromising therapeutic outcomes. Herein, pyropheophorbide-conjugated lipid (pyropheophorbide-lipid, PL) and trastuzumab were fabricated into targeted nanoparticles (TP NPs) for loading perfluorobromooctane (PFOB) carrying oxygen (TPPO NPs), thus enabling oxygen self-supplied sonodynamic and antibody therapies. In vitro experiments showed that antibody targeting significantly increased the cellular uptake of sonosensitizers, and the controlled release of oxygen was dependent on ultrasound parameters, greatly enhancing the killing effects of SDT and antibody therapy. In vivo animal experiments showed that TPPO NPs-mediated enhanced permeation and retention (EPR) effects, along with antibody targeting, improved the enrichment of sonosensitizers in tumors. Notably, ultrasound-triggered topical delivery of oxygen significantly alleviated tumor hypoxia and further improved the efficacy of SDT and antibody therapy. Given the good biosafety profile of TPPO NPs, this system holds great promise for future clinical applications in gastric cancer.
Collapse
Affiliation(s)
- Xinxin Xie
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing 100191, China
| | - Jinxia Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Lihong Sun
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Shuyu Xu
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Shiti Sha Ma
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Haonan Wang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Xiaoda Li
- Peking University Health Science Center, Beijing 100191, China
| | - Qiong Xiang
- Institute of Medicine, Medical Research Center, Jishou University, Jishou, Hunan, China
| | - Ligang Cui
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China.
| | - Xiaolong Liang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
2
|
Elshazly AM, Elzahed AA, Gewirtz DA. Evidence for cytoprotective autophagy in response to HER2-targeted monoclonal antibodies. J Pharmacol Exp Ther 2025; 392:100007. [PMID: 39892993 DOI: 10.1124/jpet.123.002048] [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: 11/30/2023] [Revised: 02/03/2024] [Accepted: 02/11/2024] [Indexed: 03/09/2024] Open
Abstract
The advent of HER2-targeted monoclonal antibodies such as trastuzumab has significantly improved the clinical outcomes for patients with breast cancer overexpressing HER2 and, more recently, also for gastric cancers. However, the development of resistance, as is frequently the case for other antineoplastic modalities, constrains their clinical efficacy. Multiple molecular mechanisms and signaling pathways have been investigated for their potential involvement in the development of resistance to HER2-targeted therapies, among which is autophagy. Autophagy is an inherent cellular mechanism whereby cytoplasmic components are selectively degraded to maintain cellular homeostasis via the generation of energy and metabolic intermediates. Although the cytoprotective form of autophagy is thought to predominate, other forms of autophagy have also been identified in response to chemotherapeutic agents in various tumor models; these include cytotoxic, cytostatic, and nonprotective functional forms of autophagy. In this review, we provide an overview of the autophagic machinery induced in response to HER2-targeted monoclonal antibodies, with a focus on trastuzumab and trastuzumab-emtansine, in an effort to determine whether autophagy targeting or modulation could be translated clinically to increase their effectiveness and/or overcome the development of resistance. SIGNIFICANCE STATEMENT: This manuscript is one in a series of papers that interrogate the role(s) of the autophagy induced in response to antineoplastic agents in various cancer models. This series of papers was developed in an effort to establish whether autophagy targeting or modulation is likely to be an effective adjuvant strategy to increase the efficacy of cancer chemotherapeutic agents. This review explores the relationship between the autophagic machinery and HER2-targeted therapies.
Collapse
Affiliation(s)
- Ahmed M Elshazly
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Aya A Elzahed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
3
|
Zhu B, Xiang K, Li T, Li X, Shi F. The signature of extracellular vesicles in hypoxic breast cancer and their therapeutic engineering. Cell Commun Signal 2024; 22:512. [PMID: 39434182 PMCID: PMC11492701 DOI: 10.1186/s12964-024-01870-w] [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: 07/03/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024] Open
Abstract
Breast cancer (BC) currently ranks second in the global cancer incidence rate. Hypoxia is a common phenomenon in BC. Under hypoxic conditions, cells in the tumor microenvironment (TME) secrete numerous extracellular vesicles (EVs) to achieve intercellular communication and alter the metabolism of primary and metastatic tumors that shape the TME. In addition, emerging studies have indicated that hypoxia can promote resistance to tumor treatment. Engineered EVs are expected to become carriers for cancer treatment due to their high biocompatibility, low immunogenicity, high drug delivery efficiency, and ease of modification. In this review, we summarize the mechanisms of EVs in the primary TME and distant metastasis of BC under hypoxic conditions. Additionally, we highlight the potential applications of engineered EVs in mitigating the malignant phenotypes of BC cells under hypoxia.
Collapse
Affiliation(s)
- Baiheng Zhu
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Kehao Xiang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Tanghua Li
- The First Clinical Medical School, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xin Li
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Fujun Shi
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
4
|
Li Y, Yu H, Han X, Pan Y. Analyses of hypoxia-related risk factors and clinical relevance in breast cancer. Front Oncol 2024; 14:1350426. [PMID: 38500661 PMCID: PMC10946248 DOI: 10.3389/fonc.2024.1350426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/12/2024] [Indexed: 03/20/2024] Open
Abstract
Introduction Hypoxia plays an important role in the heterogeneity, relapse, metastasis, and drug resistance of breast cancer. In this study, we explored the hypoxia-related biological signatures in different subtypes of breast cancer and identified the key prognostic factors by bioinformatics methods. Methods Based on The Cancer Genome Atlas (TCGA) Breast Cancer datasets, we divided the samples into immune-activated/suppressed populations by single-sample gene set enrichment analysis (ssGSEA) and then used hierarchical clustering to further identify hypoxic/non-hypoxic populations from the immune-suppressed samples. A hypoxia related risk model of breast cancer was constructed. Results Nuclear factor interleukin-3 regulated (NFIL3), serpin family E member 1 (SERPINE1), FOS, biglycan (BGN), epidermal growth factor receptor (EGFR), and sushi-repeat-containing protein, X-linked (SRPX) were identified as key hypoxia-related genes. Margin status, American Joint Committee on Cancer (AJCC) stage, hypoxia status, estrogen receptor/progesterone receptor (ER/PR) status, NFIL3, SERPINE1, EGFR, and risk score were identified as independent prognostic indicators for breast cancer patients. The 3- and 5-year survival curves of the model and immunohistochemical staining on the breast cancer microarray verified the statistical significance and feasibility of our model. Among the different molecular types of breast cancer, ER/PR+ and HER2+ patients might have higher hypoxia-related risk scores. ER/PR-negative samples demonstrated more activated immune-related pathways and better response to most anticancer agents. Discussion Our study revealed a novel risk model and potential feasible prognostic factors for breast cancer and might provide new perspectives for individual breast cancer treatment.
Collapse
Affiliation(s)
- Yan Li
- Department of Clinical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Haiyang Yu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xinghua Han
- Department of Clinical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yueyin Pan
- Department of Clinical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
5
|
Kotb RM, Ibrahim SS, Mostafa OM, Shahin NN. Potential role of CXCR4 in trastuzumab resistance in breast cancer patients. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166520. [PMID: 35985446 DOI: 10.1016/j.bbadis.2022.166520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/07/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022]
Abstract
Despite the efficacy of trastuzumab in treating HER2-positive breast cancer patients, a significant proportion of patients relapse after treatment. The role of C-X-C chemokine receptor type 4 (CXCR4) in trastuzumab resistance was studied only in cell lines and the underlying mechanisms remain largely unclear. This study investigated the role of CXCR4 in trastuzumab resistance in breast cancer patients and explored the possible underlying mechanisms. The study was performed retrospectively on tissue samples from 62 breast cancer patients including 42 who were treated with trastuzumab and chemotherapy and 20 who received chemotherapy alone in adjuvant setting. Expression levels of CXCR4 and its regulators hypoxia-inducible factor 1-alpha (HIF-1α), tristetraprolin (TTP), human antigen R (HuR), itchy E3 ubiquitin protein ligase (ITCH), miR-302a and miR-494 were determined and their associations with tumor recurrence and disease-free survival were analyzed. In trastuzumab-treated patients, high CXCR4 expression was associated with recurrence and was an independent predictor of progression risk after therapy. CXCR4 correlated positively with its transcriptional regulator, HIF-1α, and negatively with its post-translational regulator, ITCH. HIF-1α, HuR and ITCH were significantly associated with clinical outcome. In chemotherapy-treated patients, neither CXCR4 nor any of its regulators were associated with recurrence or predicted disease progression risk after chemotherapy. In conclusion, this study suggests a potential role for CXCR4 in recurrence after trastuzumab-based therapy in human breast cancer that could be mediated, at least in part, by hypoxia and/or decreased ubiquitination. These findings highlight the potential utility of CXCR4 as a promising target for enhancing trastuzumab therapeutic outcome.
Collapse
Affiliation(s)
- Ranim M Kotb
- General Administration of Clinical Trials, Central Administration of Biological and Innovative Products and Clinical Studies, Egyptian Drug Authority, Giza, Egypt
| | - Safinaz S Ibrahim
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Osama M Mostafa
- Department of Pathology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Nancy N Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| |
Collapse
|
6
|
Hao J, Yu L, Lu H, Sakthivel TS, Seal S, Liu B, Zhao J. Sensitization of breast cancer to Herceptin by redox active nanoparticles. Am J Cancer Res 2021; 11:4884-4899. [PMID: 34765298 PMCID: PMC8569362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/30/2021] [Indexed: 06/13/2023] Open
Abstract
Herceptin-resistant tumor relapse remains a major clinical issue responsible for the poor prognosis of HER2+ breast cancer. Understanding the underlying mechanisms and finding a therapeutic solution are of paramount urgency to improve the patient management. Here we report that anticancer redox active cerium oxide nanoparticles (CONPs) can potently sensitize the cancer cells to the cytotoxicity of Herceptin. By comparing between Herceptin-sensitive and Herceptin-resistant human breast cancer cell lines under normoxic as well as hypoxic culture conditions, we found that in the presence of CONPs, Herceptin can kill the Herceptin-resistant cells equally effectively as it kills the Herceptin-sensitive cells under the hypoxic, but not normoxic, culture conditions by inhibiting the cell viability, survival and proliferation. Signaling analysis reveals that under the normoxic conditions, the levels of hypoxia induced factor 1α as well as vascular endothelial growth factor are higher in the Herceptin-resistant cells than that in the Herceptin-sensitive cells and are strongly induced once the culture is switched to the hypoxic conditions, which can be potently suppressed by CONPs. Treatment with CONPs plus Herceptin significantly slows down the primary tumor growth and lung metastasis of the Herceptin-resistant cells in a xenograft mouse model of orthotopic breast cancer through inhibiting the cell proliferation and survival as well as tumor angiogenesis. These results shed new lights on the mechanisms underlying the Herceptin resistance of the HER2+ breast cancer and provide insights into introducing CONPs-like agents to Herceptin-based therapy to improve treatment outcomes.
Collapse
Affiliation(s)
- Jie Hao
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, OrlandoFL 32827, USA
| | - Lin Yu
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, OrlandoFL 32827, USA
| | - Heng Lu
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, OrlandoFL 32827, USA
- University of Miami Miller School of MedicineMiami, FL 33136, USA
| | - Tamil S Sakthivel
- Department of Material Science and Engineering, Advanced Materials Processing and Analysis Center, Nanoscience Technology Center, Biionix Cluster, College of Medicine, University of Central FloridaOrlando, FL 32816, USA
| | - Sudipta Seal
- Department of Material Science and Engineering, Advanced Materials Processing and Analysis Center, Nanoscience Technology Center, Biionix Cluster, College of Medicine, University of Central FloridaOrlando, FL 32816, USA
| | - Bolin Liu
- Department of Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences CenterNew Orleans, LA 70112, USA
| | - Jihe Zhao
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, OrlandoFL 32827, USA
| |
Collapse
|
7
|
Du JX, Chen C, Luo YH, Cai JL, Cai CZ, Xu J, Ni XJ, Zhu W. Establishment and validation of a novel autophagy-related gene signature for patients with breast cancer. Gene 2020; 762:144974. [PMID: 32707305 DOI: 10.1016/j.gene.2020.144974] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 07/17/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND There exists considerable evidence conforming that autophagy may play an important role in the biological process of breast cancer. This study aimed to construct and evaluate a novel autophagy-related gene signature as a potential prognostic factor and therapeutic target in breast cancer patients based on high-throughput sequencing datasets. MATERIALS & METHODS Autophagy-related genes obtained from the Human Autophagy Database and high-sequencing data obtained from The Cancer Genome Atlas (TCGA) were analyzed to identify differential expressed genes (DEGs) between tumor and normal tissues. Then GO and KEGG analysis were performed to explore potential biological and pathological functions of DEGs. Autophagy-related prognostic genes were identified by univariate COX regression analysis. Subsequently stepwise model selection using the Alkaike information criterion (AIC) and multivariate COX regression model was performed to construct autophagy-related gene signature. Then patients were divided into high- and low-risk groups based on the risk score identified by the autophagy-related gene signature. Multivariate COX regression model and stratification analysis were used to specify the prognostic value of this gene signature in whole cohort and various subgroups. T-test and ANOVA analysis were used to compare the expression differences of continuous variables (5 prognostic genes and risk score) in binary and multiple category groups respectively. Kaplan-Meier analysis, log-rank tests and the area under receiver operating characteristic (ROC) curve (AUC) were conducted to validate the accuracy and precise of the autophagy-related gene signature based on GSE20685 and GSE21653 datasets. RESULTS We profiled autophagy-related DEGs in normal and breast tumor tissues. GO and KEGG analysis indicated that autophagy-related DEGs might participate in breast cancer occurrence, development and drug resistance. Then we identified five autophagy-related genes (EIF4EBP1, ATG4A, BAG1, MAP1LC3A and SERPINA1) that had significantly prognostic values for breast cancer. Autophagy-related gene signature was constructed and patients were divided into high- and low- risk groups based on their risk score. Patients in the high-risk group tended to have shorter overall survival (OS) and relapse-free survival (RFS) times than those in the low-risk group (OS: HR = 1.620, 95%CIs: 1.345-1.950; P < 0.001; RFS: HR = 1.487, 95%CIs: 1.248-1.771, P < 0.001). Autophagy-related gene signature had significant prognostic value in stratified subgroups especially in advanced breast cancer subgroups (T3-4; N2-3; stage III-IV). Its prognostic value was further confirmed in two GEO validation datasets (GSE20685: P = 6.795e-03; GSE21653: P = 1.383e-03). Finally, association analysis between clinicopathological factors and gene signature showed the risk score was higher in patients with ER/PR negative, higher clinical stage or T stage (P < 0.01). CONCLUSION We established and confirmed a novel autophagy-related gene signature for patients with breast cancer that had independent survival prognostic value especially in advanced breast cancer subgroups. Our research might promote the molecular mechanism study of autophagy-related genes in breast cancer.
Collapse
Affiliation(s)
- Jun-Xian Du
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cong Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi-Hong Luo
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jia-Liang Cai
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200032, China
| | - Cheng-Zhe Cai
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jing Xu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiao-Jian Ni
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Wei Zhu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
8
|
de Heer EC, Jalving M, Harris AL. HIFs, angiogenesis, and metabolism: elusive enemies in breast cancer. J Clin Invest 2020; 130:5074-5087. [PMID: 32870818 PMCID: PMC7524491 DOI: 10.1172/jci137552] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hypoxia-inducible factors (HIFs) and the HIF-dependent cancer hallmarks angiogenesis and metabolic rewiring are well-established drivers of breast cancer aggressiveness, therapy resistance, and poor prognosis. Targeting of HIF and its downstream targets in angiogenesis and metabolism has been unsuccessful so far in the breast cancer clinical setting, with major unresolved challenges residing in target selection, development of robust biomarkers for response prediction, and understanding and harnessing of escape mechanisms. This Review discusses the pathophysiological role of HIFs, angiogenesis, and metabolism in breast cancer and the challenges of targeting these features in patients with breast cancer. Rational therapeutic combinations, especially with immunotherapy and endocrine therapy, seem most promising in the clinical exploitation of the intricate interplay of HIFs, angiogenesis, and metabolism in breast cancer cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Ellen C. de Heer
- University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Groningen, Netherlands
| | - Mathilde Jalving
- University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Groningen, Netherlands
| | - Adrian L. Harris
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
9
|
Rybinska I, Sandri M, Bianchi F, Orlandi R, De Cecco L, Gasparini P, Campiglio M, Paolini B, Sfondrini L, Tagliabue E, Triulzi T. Extracellular Matrix Features Discriminate Aggressive HER2-Positive Breast Cancer Patients Who Benefit from Trastuzumab Treatment. Cells 2020; 9:cells9020434. [PMID: 32069815 PMCID: PMC7072535 DOI: 10.3390/cells9020434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/11/2022] Open
Abstract
We previously identified an extracellular matrix (ECM) gene expression pattern in breast cancer (BC), called ECM3, characterized by a high expression of genes encoding structural ECM proteins. Since ECM is reportedly implicated in response to therapy of BCs, the aim of this work is to investigate the prognostic and predictive value of ECM3 molecular classification in HER2-positive BCs. ECM3 resulted in a robust cluster that identified a subset of 25-37% of HER2-positive tumors with molecular aggressive features. ECM3 was significantly associated with worse prognosis in two datasets of HER2-positive BCs untreated with adjuvant therapy. Analyses carried out on two of our cohorts of patients treated or not with adjuvant trastuzumab showed association of ECM3 with worse prognosis only in patients not treated with trastuzumab. Moreover, investigating a dataset that includes gene profile data of tumors treated with neoadjuvant trastuzumab plus chemotherapy or chemotherapy alone, ECM3 was associated with increased pathological complete response if treated with trastuzumab. In the in vivo experiments, increased diffusion and trastuzumab activity were found in tumors derived from injection of HER2-positive cells with Matrigel that creates an ECM-rich tumor environment. Taken together, these results indicate that HER2-positive BCs classified as ECM3 have an aggressive phenotype but they are sensitive to trastuzumab treatment.
Collapse
Affiliation(s)
- Ilona Rybinska
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (F.B.); (R.O.); (M.C.); (T.T.)
| | - Marco Sandri
- Data Methods and Systems Statistical Laboratory, University of Brescia, 25121 Brescia, Italy;
| | - Francesca Bianchi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (F.B.); (R.O.); (M.C.); (T.T.)
| | - Rosaria Orlandi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (F.B.); (R.O.); (M.C.); (T.T.)
| | - Loris De Cecco
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Patrizia Gasparini
- Genomic Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Manuela Campiglio
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (F.B.); (R.O.); (M.C.); (T.T.)
| | - Biagio Paolini
- Anatomic Pathology A Unit, Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (F.B.); (R.O.); (M.C.); (T.T.)
- Correspondence:
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (F.B.); (R.O.); (M.C.); (T.T.)
| |
Collapse
|
10
|
Triulzi T, Regondi V, De Cecco L, Cappelletti MR, Di Modica M, Paolini B, Lollini PL, Di Cosimo S, Sfondrini L, Generali D, Tagliabue E. Early immune modulation by single-agent trastuzumab as a marker of trastuzumab benefit. Br J Cancer 2018; 119:1487-1494. [PMID: 30478407 PMCID: PMC6288086 DOI: 10.1038/s41416-018-0318-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Optimising the selection of HER2-targeted regimens by identifying subsets of HER2-positive breast cancer (BC) patients who need more or less therapy remains challenging. We analysed BC samples before and after treatment with 1 cycle of trastuzumab according to the response to trastuzumab. METHODS Gene expression profiles of pre- and post-treatment tumour samples from 17 HER2-positive BC patients were analysed on the Illumina platform. Tumour-associated immune pathways and blood counts were analysed with regard to the response to trastuzumab. HER2-positive murine models with differential responses to trastuzumab were used to reproduce and better characterise these data. RESULTS Patients who responded to single-agent trastuzumab had basal tumour biopsies that were enriched in immune pathways, particularly the MHC-II metagene. One cycle of trastuzumab modulated the expression levels of MHC-II genes, which increased in patients who had a complete response on treatment with trastuzumab and chemotherapy. Trastuzumab increased the MHC-II-positive cell population, primarily macrophages, only in the tumour microenvironment of responsive mice. In patients who benefited from complete trastuzumab therapy and in mice that harboured responsive tumours circulating neutrophil levels declined, but this cell subset rose in nonresponsive tumours. CONCLUSIONS Short treatment with trastuzumab induces local and systemic immunomodulation that is associated with clinical outcomes.
Collapse
Affiliation(s)
- Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Viola Regondi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Loris De Cecco
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Maria Rosa Cappelletti
- U.O. Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, Azienda Socio-Sanitaria Territoriale di Cremona, Cremona, Italy
| | - Martina Di Modica
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Biagio Paolini
- Anatomic Pathology A Unit, Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Pier Luigi Lollini
- Laboratory of Immunology and Biology of Metastases, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Serena Di Cosimo
- Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milano, Italy
| | - Daniele Generali
- U.O. Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, Azienda Socio-Sanitaria Territoriale di Cremona, Cremona, Italy
- Dipartimento Universitario Clinico di Scienze Mediche, Chirurgiche e della Salute, Università degli Studi di Trieste, Trieste, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy.
| |
Collapse
|
11
|
Can hi-jacking hypoxia inhibit extracellular vesicles in cancer? Drug Discov Today 2018; 23:1267-1273. [PMID: 29577970 DOI: 10.1016/j.drudis.2018.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/15/2018] [Accepted: 03/14/2018] [Indexed: 12/11/2022]
Abstract
Increasing evidence indicates that extracellular vesicles (EVs) are key players in undesirable cell-cell communication in cancer. However, the release of EVs is not unique to cancer cells; normal cells release EVs to perform physiological roles. Thus, selective inhibition of EV release from cancer cells is desirable. Hypoxia contributes to tumour development and aggressiveness. EV quantities and thus undesirable communications are substantially increased in hypoxia. Targeting hypoxia could selectively inhibit EV release from tumour cells without disturbing physiologically relevant EVs. The unfavourable association between hypoxia and EV release is evident in multiple tumour types; therefore, targeting hypoxia could have a broad therapeutic benefit.
Collapse
|
12
|
Rodríguez CE, Berardi DE, Abrigo M, Todaro LB, Bal de Kier Joffé ED, Fiszman GL. Breast cancer stem cells are involved in Trastuzumab resistance through the HER2 modulation in 3D culture. J Cell Biochem 2017; 119:1381-1391. [DOI: 10.1002/jcb.26298] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/18/2017] [Indexed: 01/15/2023]
Affiliation(s)
- Cristina E. Rodríguez
- Universidad de Buenos Aires, Área Investigación, Instituto de Oncología Ángel H. RoffoBuenos AiresArgentina
| | - Damian E. Berardi
- Universidad de Buenos Aires, Área Investigación, Instituto de Oncología Ángel H. RoffoBuenos AiresArgentina
| | - Marianela Abrigo
- Universidad de Buenos Aires, Área Investigación, Instituto de Oncología Ángel H. RoffoBuenos AiresArgentina
| | - Laura B. Todaro
- Universidad de Buenos Aires, Área Investigación, Instituto de Oncología Ángel H. RoffoBuenos AiresArgentina
| | - Elisa D. Bal de Kier Joffé
- Universidad de Buenos Aires, Área Investigación, Instituto de Oncología Ángel H. RoffoBuenos AiresArgentina
| | - Gabriel L. Fiszman
- Universidad de Buenos Aires, Área Investigación, Instituto de Oncología Ángel H. RoffoBuenos AiresArgentina
| |
Collapse
|
13
|
Zhang L, Huang Y, Zhuo W, Zhu Y, Zhu B, Chen Z. Identification and characterization of biomarkers and their functions for Lapatinib-resistant breast cancer. Med Oncol 2017; 34:89. [DOI: 10.1007/s12032-017-0953-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 04/05/2017] [Indexed: 11/29/2022]
|
14
|
Di Modica M, Sfondrini L, Regondi V, Varchetta S, Oliviero B, Mariani G, Bianchi GV, Generali D, Balsari A, Triulzi T, Tagliabue E. Taxanes enhance trastuzumab-mediated ADCC on tumor cells through NKG2D-mediated NK cell recognition. Oncotarget 2016; 7:255-65. [PMID: 26595802 PMCID: PMC4807996 DOI: 10.18632/oncotarget.6353] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/09/2015] [Indexed: 01/14/2023] Open
Abstract
Recent clinical data indicate a synergistic therapeutic effect between trastuzumab and taxanes in neoadjuvantly treated HER2-positive breast cancer (BC) patients. In HER2+ BC experimental models and patients, we investigated whether this synergy depends on the ability of drug-induced stress to improve NK cell effectiveness and thus trastuzumab-mediated ADCC. HER2+ BC cell lines BT474 and MDAMB361 treated with docetaxel showed up-modulation of NK activator ligands both in vitro and in vivo, accompanied by a 15-40% increase in in vitro trastuzumab-mediated ADCC; antibodies blocking the NKG2D receptor significantly reduced this enhancement. NKG2D receptor expression was increased by docetaxel treatment in circulating and splenic NK cells from mice xenografted with tumor cells, an increase related to expansion of the CD11b+Ly6G+ cell population. Accordingly, NK cells derived from HER2+ BC patients after treatment with taxane-containing therapy expressed higher levels of NKG2D receptor than before treatment. Moreover, plasma obtained from these patients recapitulated the modulation of NKG2D on healthy donors' NK cells, improving their trastuzumab-mediated activity in vitro. This enhancement occurred mainly using plasma from patients with low NKG2D basal expression. Our results indicate that taxanes increase tumor susceptibility to ADCC by acting on tumor and NK cells, and suggest that taxanes concomitantly administered with trastuzumab could maximize the antibody effect, especially in patients with low basal immune effector cytotoxic activity.
Collapse
Affiliation(s)
- Martina Di Modica
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Viola Regondi
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Stefania Varchetta
- Department of Infectious Diseases, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Barbara Oliviero
- Department of Infectious Diseases, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Gabriella Mariani
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Valeria Bianchi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Daniele Generali
- Dipartimento di Terapia Molecolare e Farmacogenomica, Istituti Ospitalieri di Cremona, Cremona, Italy
| | - Andrea Balsari
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
15
|
Roviello G, Milani M, Gobbi A, Dester M, Cappelletti MR, Allevi G, Aguggini S, Ravelli A, Gussago F, Cocconi A, Zanotti L, Senti C, Strina C, Bottini A, Generali D. A Phase II study of olaparib in breast cancer patients: biological evaluation from a 'window of opportunity' trial. Future Oncol 2016; 12:2189-2193. [PMID: 27324108 DOI: 10.2217/fon-2016-0116] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/26/2016] [Indexed: 11/21/2022] Open
Abstract
The OLTRE trial (ClinicalTrials.gov number: NCT02681562) is an open-label, 'window of opportunity' Phase II controlled trial to evaluate the biological activity of olaparib in locally advanced triple-negative breast cancer compared with other subtypes of locally advanced breast cancer patients carrying germinal BRCA mutation receiving olaparib with the same treatment approach. The primary end point is to investigate the correlation between baseline gene and protein expression profile in order to identify possible predictive markers of response to olaparib. The OLTRE trial is expected to identify the surrogate markers of the biological activity of olaparib in the treatment of patients with triple-negative breast cancer.
Collapse
Affiliation(s)
- Giandomenico Roviello
- Section of Pharmacology & University Center DIFF-Drug Innovation Forward Future, Department of Molecular & Translational Medicine, University of Brescia, Viale Europa 11, 25124 Brescia, Italy
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | - Manuela Milani
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | - Angela Gobbi
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | - Martina Dester
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | | | - Giovanni Allevi
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | - Sergio Aguggini
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | - Andrea Ravelli
- Section of Experimental Oncology, Department of Clinical & Experimental Medicine, University of Parma, Via Università, 12, 43121 Parma, Italy
| | - Francesca Gussago
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | | | - Laura Zanotti
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | - Chiara Senti
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | - Carla Strina
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | - Alberto Bottini
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | - Daniele Generali
- US Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129 Trieste, Italy
| |
Collapse
|
16
|
Triulzi T, De Cecco L, Sandri M, Prat A, Giussani M, Paolini B, Carcangiu ML, Canevari S, Bottini A, Balsari A, Menard S, Generali D, Campiglio M, Di Cosimo S, Tagliabue E. Whole-transcriptome analysis links trastuzumab sensitivity of breast tumors to both HER2 dependence and immune cell infiltration. Oncotarget 2016; 6:28173-82. [PMID: 26334217 PMCID: PMC4695052 DOI: 10.18632/oncotarget.4405] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/19/2015] [Indexed: 01/21/2023] Open
Abstract
While results thus far demonstrate the clinical benefit of trastuzumab, some patients do not respond to this therapy. To identify a molecular predictor of trastuzumab benefit, we conducted whole-transcriptome analysis of primary HER2+ breast carcinomas obtained from patients treated with trastuzumab-containing therapies and correlated the molecular portrait with treatment benefit. The estimated association between gene expression and relapse-free survival allowed development of a trastuzumab risk model (TRAR), with ERBB2 and ESR1 expression as core elements, able to identify patients with high and low risk of relapse. Application of the TRAR model to 24 HER2+ core biopsies from patients treated with neo-adjuvant trastuzumab indicated that it is predictive of trastuzumab response. Examination of TRAR in available whole-transcriptome datasets indicated that this model stratifies patients according to response to trastuzumab-based neo-adjuvant treatment but not to chemotherapy alone. Pathway analysis revealed that TRAR-low tumors expressed genes of the immune response, with higher numbers of CD8-positive cells detected immunohistochemically compared to TRAR-high tumors. The TRAR model identifies tumors that benefit from trastuzumab-based treatment as those most enriched in CD8-positive immune infiltrating cells and with high ERBB2 and low ESR1 mRNA levels, indicating the requirement for both features in achieving trastuzumab response.
Collapse
Affiliation(s)
- Tiziana Triulzi
- Department of Experimental Oncology and Molecular Medicine, Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Loris De Cecco
- Department of Experimental Oncology and Molecular Medicine, Functional Genomics Core Facility, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco Sandri
- Department of Experimental Oncology and Molecular Medicine, Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Aleix Prat
- Translational Genomics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Medical Oncology Department, Hospital Clínic i Provincial, Barcelona, Spain
| | - Marta Giussani
- Department of Experimental Oncology and Molecular Medicine, Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Biagio Paolini
- Department of Pathology, Anatomic Pathology A Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marialuisa L Carcangiu
- Department of Pathology, Anatomic Pathology A Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Canevari
- Department of Experimental Oncology and Molecular Medicine, Functional Genomics Core Facility, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alberto Bottini
- Dipartimento di Terapia Molecolare e Farmacogenomica, Istituti Ospitalieri di Cremona, Italy
| | - Andrea Balsari
- Department of Experimental Oncology and Molecular Medicine, Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Italy
| | - Sylvie Menard
- Department of Experimental Oncology and Molecular Medicine, Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Daniele Generali
- Dipartimento di Terapia Molecolare e Farmacogenomica, Istituti Ospitalieri di Cremona, Italy
| | - Manuela Campiglio
- Department of Experimental Oncology and Molecular Medicine, Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Serena Di Cosimo
- Department of Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elda Tagliabue
- Department of Experimental Oncology and Molecular Medicine, Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
17
|
Liu M, Li Z, Yang J, Jiang Y, Chen Z, Ali Z, He N, Wang Z. Cell-specific biomarkers and targeted biopharmaceuticals for breast cancer treatment. Cell Prolif 2016; 49:409-20. [PMID: 27312135 PMCID: PMC6496337 DOI: 10.1111/cpr.12266] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 05/04/2016] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is the second leading cause of cancer death among women, and its related treatment has been attracting significant attention over the past decades. Among the various treatments, targeted therapy has shown great promise as a precision treatment, by binding to cancer cell-specific biomarkers. So far, great achievements have been made in targeted therapy of breast cancer. In this review, we first discuss cell-specific biomarkers, which are not only useful for classification of breast cancer subtyping but also can be utilized as goals for targeted therapy. Then, the innovative and generic-targeted biopharmaceuticals for breast cancer, including monoclonal antibodies, non-antibody proteins and small molecule drugs, are reviewed. Finally, we provide our outlook on future developments of biopharmaceuticals, and provide solutions to problems in this field.
Collapse
Affiliation(s)
- Mei Liu
- School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Zhiyang Li
- School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
- Department of Laboratory MedicineNanjing Drum Tower Hospital Clinical CollegeNanjing UniversityNanjingChina
| | - Jingjing Yang
- School of Chemistry and Chemical EngineeringSoutheast UniversityNanjingChina
| | - Yanyun Jiang
- School of Chemistry and Chemical EngineeringSoutheast UniversityNanjingChina
| | - Zhongsi Chen
- School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Zeeshan Ali
- School of Chemistry and Chemical EngineeringSoutheast UniversityNanjingChina
| | - Nongyue He
- School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Zhifei Wang
- School of Chemistry and Chemical EngineeringSoutheast UniversityNanjingChina
| |
Collapse
|
18
|
Koukourakis MI, Kalamida D, Mitrakas A, Pouliliou S, Kalamida S, Sivridis E, Giatromanolaki A. Intensified autophagy compromises the efficacy of radiotherapy against prostate cancer. Biochem Biophys Res Commun 2015; 461:268-74. [PMID: 25887800 DOI: 10.1016/j.bbrc.2015.04.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 04/04/2015] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Radiotherapy is an equivalent alternative or complement to radical prostatectomy, with high therapeutic efficacy. High risk patients, however, experience high relapse rates, so that research on radio-sensitization is the most evident route to improve curability of this common disease. MATERIALS AND METHODS In the current study we investigated the autophagic activity in a series of patients with localized prostate tumors treated with radical radiotherapy, using the LC3A and the LAMP2a proteins as markers of autophagosome and lysosome cellular content, respectively. The role of autophagy on prostate cancer cell line resistance to radiation was also examined. RESULTS Using confocal microscopy on tissue biopsies, we showed that prostate cancer cells have, overall, high levels of LC3A and low levels of LAMP2a compared to normal prostate glands. Tumors with a 'highLC3A/lowLAMP2a' phenotype, suggestive of intensified lysosomal consumption, had a significantly poorer biochemical relapse free survival. The PC3 radioresistant cell line sustained remarkably its autophagic flux ability after radiation, while the DU145 radiosensitive one experiences a prolonged blockage of the autophagic process. This was assessed with aggresome accumulation detection and LC3A/LAMP2a double immunofluorescence, as well as with sequestrosome/p62 protein detection. By silencing the LC3A or LAMP2a expression, both cell lines became more sensitive to escalated doses of radiation. CONCLUSIONS High base line autophagy activity and cell ability to sustain functional autophagy define resistance of prostate cancer cells to radiotherapy. This can be reversed by blocking up-regulated components of the autophagy pathway, which may prove of importance in the field of clinical radiotherapy.
Collapse
Affiliation(s)
- Michael I Koukourakis
- Department of Radiotherapy/Oncology, Democritus University of Thrace, Alexandroupolis, 68100, Greece.
| | - Dimitra Kalamida
- Department of Radiotherapy/Oncology, Democritus University of Thrace, Alexandroupolis, 68100, Greece
| | - Achilleas Mitrakas
- Department of Radiotherapy/Oncology, Democritus University of Thrace, Alexandroupolis, 68100, Greece
| | - Stamatia Pouliliou
- Department of Radiotherapy/Oncology, Democritus University of Thrace, Alexandroupolis, 68100, Greece
| | - Sofia Kalamida
- Department of Radiotherapy/Oncology, Democritus University of Thrace, Alexandroupolis, 68100, Greece
| | - Efthimios Sivridis
- Department of Pathology, Democritus University of Thrace, Alexandroupolis, 68100, Greece
| | | |
Collapse
|