1
|
Hao C, Wang X, Shi Y, Tong Z, Li S, Liu X, Zhang L, Zhang J, Meng W, Zhang L. Combination Therapy of Pyrotinib and Metronomic Vinorelbine in HER2+ Advanced Breast Cancer after Trastuzumab Failure (PROVE): A Prospective Phase 2 Study. Cancer Res Treat 2025; 57:434-442. [PMID: 39118524 PMCID: PMC12016823 DOI: 10.4143/crt.2024.340] [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: 04/03/2024] [Accepted: 08/08/2024] [Indexed: 08/10/2024] Open
Abstract
PURPOSE Approximately 50%-74% of patients with metastatic human epidermal growth factor receptor 2 (HER2)-positive breast cancer do not respond to trastuzumab, with 75% of treated patients experiencing disease progression within a year. The combination of pyrotinib and capecitabine has showed efficacy in these patients. This study evaluates the efficacy and safety of pyrotinib combined with metronomic vinorelbine for trastuzumab-pretreated HER2-positive advanced breast cancer patients. MATERIALS AND METHODS In this phase 2 trial, patients aged 18-75 years with HER2-positive advanced breast cancer who had previously failed trastuzumab treatment were enrolled to receive pyrotinib 400 mg daily in combination with vinorelbine 40mg thrice weekly. The primary endpoint was progression-free survival (PFS), while secondary endpoints included objective response rate (ORR), disease control rate (DCR), overall survival (OS), and safety. RESULTS From October 21, 2019, to January 21, 2022, 36 patients were enrolled and received at least one dose of study treatment. At the cutoff date, 20 experienced disease progression or death. With a median follow-up duration of 35 months, the median PFS was 13.5 months (95% confidence interval [CI], 8.3 to 18.5). With all patients evaluated, an ORR of 38.9% (95% CI, 23.1 to 56.5) and a DCR of 83.3% (95% CI, 67.2 to 93.6) were achieved. The median OS was not reached. Grade 3 adverse events (AEs) were observed in 17 patients, with diarrhea being the most common (27.8%), followed by vomiting (8.3%) and stomachache (5.6%). There were no grade 4/5 AEs. CONCLUSION Pyrotinib combined with metronomic vinorelbine showed promising efficacy and an acceptable safety profile in HER2-positive advanced breast cancer patients after trastuzumab failure.
Collapse
Affiliation(s)
- Chunfang Hao
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Department of Breast Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Xu Wang
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Yehui Shi
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Zhongsheng Tong
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Shufen Li
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Xiaodong Liu
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Lan Zhang
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Jie Zhang
- Department of Breast Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Wenjing Meng
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Li Zhang
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
2
|
Liu D, Jee J, Drilon A, Heilmann AM, Allen JM, Schrock AB, Keller-Evans RB, Li BT. Diverse ERBB2/ERBB3 Activating Alterations and Coalterations Have Implications for HER2/3-Targeted Therapies across Solid Tumors. CANCER RESEARCH COMMUNICATIONS 2025; 5:680-693. [PMID: 40178042 PMCID: PMC12022956 DOI: 10.1158/2767-9764.crc-24-0620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/06/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
SIGNIFICANCE CGP provides genomic context for HER2 status beyond the information provided by IHC and FISH, including detection of ERBB2 mutations and co-alterations that may suggest sensitivity/resistance to HER2-directed therapies, and is therefore crucial for guiding treatment choice and understanding individual patient response.
Collapse
Affiliation(s)
- Dazhi Liu
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justin Jee
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | | | | | | | | | - Bob T. Li
- Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
3
|
Kim Y, Song J, Kim N, Sim T. Recent progress in emerging molecular targeted therapies for intrahepatic cholangiocarcinoma. RSC Med Chem 2025:d4md00881b. [PMID: 39925737 PMCID: PMC11800140 DOI: 10.1039/d4md00881b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/11/2025] [Indexed: 02/11/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a diverse group of epithelial malignant tumors arising from the biliary tract, characterized by high molecular heterogeneity. It is classified into intrahepatic (iCCA) and extrahepatic CCA (eCCA) based on the location of the primary tumor. CCA accounts for approximately 15% of all primary liver cancers, with iCCA comprising 10-20% of all CCAs. iCCA is especially known for its characteristic aggressiveness and refractoriness, leading to poor prognosis. Despite the increasing global incidence and mortality rates, surgery remains the only available standard treatment approach for a subset (25%) of patients with early-stage, resectable iCCA. The paucity of effective systemic medical therapies restricts therapeutic options for patients with advanced or metastatic iCCA. In the past decade, advances in the understanding of the molecular complexity of these tumors have provided fruitful insights for the identification of promising new druggable targets and the development of feasible therapeutic strategies that may improve treatment outcomes for patients with iCCA. In this review, we aim to highlight critical up-to-date studies and medicinal chemistry aspects, focusing on novel targeted approaches utilizing promising candidates for molecular targeted therapy in iCCA. These candidates include aberrations in isocitrate dehydrogenase (IDH) 1/2, fibroblast growth factor receptor (FGFR), B-Raf proto-oncogene (BRAF), neurotrophic tyrosine receptor kinase (NTRK), human epidermal growth factor receptor 2 (HER2), and programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1). Furthermore, this review provides an overview of potential inhibitors aimed at overcoming acquired drug resistance in these actionable targets for iCCA.
Collapse
Affiliation(s)
- Younghoon Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea +822 2228 0797
- Clinical Candidate Discovery & Development Institute, Yonsei University College of Medicine Seoul Korea
| | - Jaewon Song
- Graduate School of Clinical Drug Discovery & Development, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea
| | - Namkyoung Kim
- Department of Biomedical Sciences, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea +822 2228 0797
| | - Taebo Sim
- KU-KIST Graduate School of Converging Science and Technology, Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea +822 2228 0797
- Clinical Candidate Discovery & Development Institute, Yonsei University College of Medicine Seoul Korea
- Graduate School of Clinical Drug Discovery & Development, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea
| |
Collapse
|
4
|
Veeraraghavan J, De Angelis C, Gutierrez C, Liao FT, Sabotta C, Rimawi MF, Osborne CK, Schiff R. HER2-Positive Breast Cancer Treatment and Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:495-525. [PMID: 39821040 DOI: 10.1007/978-3-031-70875-6_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
HER2-positive (+) breast cancer is an aggressive disease with poor prognosis, a narrative that changed drastically with the advent and approval of trastuzumab, the first humanized monoclonal antibody targeting HER2. In addition to another monoclonal antibody, more classes of HER2-targeted agents, including tyrosine kinase inhibitors, and antibody-drug conjugates were developed in the years that followed. While these potent therapies have substantially improved the outcome of patients with HER2+ breast cancer, resistance has prevailed as a clinical challenge ever since the arrival of targeted agents. Efforts to develop new treatment regimens to treat/overcome resistance is futile without a primary understanding of the mechanistic underpinnings of resistance. Resistance could be attributed to mechanisms that are either specific to the tumor epithelial cells or those that emerge through changes in the tumor microenvironment. Reactivation of the HER receptor layer due to incomplete blockade of the HER receptor layer or due to alterations in the HER receptors is one of the major mechanisms. In other instances, resistance may occur due to deregulations in key downstream signaling such as the PI3K/AKT or RAS/MEK/ERK pathways or due to the emergence of compensatory pathways such as ER, other RTKs, or metabolic pathways. Potent new targeted agents and approaches to target key actionable drivers of resistance have already been identified, many of which are in early clinical development or under preclinical evaluation. Ongoing and future translational research will continue to uncover additional therapeutic vulnerabilities, as well as new targeted agents and approaches to treat and/or overcome anti-HER2 treatment resistance.
Collapse
Affiliation(s)
- Jamunarani Veeraraghavan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Carmine De Angelis
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Carolina Gutierrez
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Fu-Tien Liao
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Caroline Sabotta
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Mothaffar F Rimawi
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - C Kent Osborne
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Rachel Schiff
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
5
|
Thornton A, Komati R, Kim H, Myers J, Petty K, Sam R, Johnson-Henderson E, Reese K, Tran L, Sridhar V, Williams C, Sridhar J. Development of 6-amido-4-aminoisoindolyn-1,3-diones as p70S6K1 inhibitors and potential breast cancer therapeutics. Front Mol Biosci 2024; 11:1481912. [PMID: 39749216 PMCID: PMC11694070 DOI: 10.3389/fmolb.2024.1481912] [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: 08/16/2024] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction Many breast cancer therapeutics target the PI3K/AKT/mTOR oncogenic pathway. Development of resistance to the therapeutics targeting this pathway is a frequent occurrence. Therapeutics targeting p70S6K1, a downstream member of this pathway, have recently gained importance due to its critical role in all types of breast cancer and its status as a prognostic marker. We have developed a new class of p70S6K1 inhibitors that show growth inhibition of MCF7 breast cancer cells. Methods A series of 6-amido-4-aminoisoindolyn-1,3-dione compounds was developed against p70S6K1 using docking, computational modeling tools, and synthesis of the designed compounds. The p70S6K1 inhibition potency of the compounds was investigated in an initial high-throughput screening followed by IC50 determination for the most active ones. The best compounds were subjected to proliferation assays on MCF7 breast cancer cells. The targeting of p70S6K1 by the compounds was confirmed by studying the phosphorylation status of downstream protein rpS6. Results In this study, we have identified a new class of compounds as p70S6K1 inhibitors that function as growth inhibitors of MCF7 breast cancer cells. The structural features imparting p70S6K1 inhibition potency to the compounds have been mapped. Our studies indicate that substitutions on the phenacetyl group residing in the cleft A of the protein do not contribute to the inhibition potency. Three compounds (5b, 5d, and 5f) have been identified to have sub-micromolar inhibition potency for p70S6K1. These compounds also exhibited growth inhibition of MCF7 cells by 40%-60% in the presence of estradiol.
Collapse
Affiliation(s)
- Adrian Thornton
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, United States
| | - Rajesh Komati
- Department of Chemistry, Nicholls State University, Thibodaux, LA, United States
| | - Hogyoung Kim
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, United States
| | - Jamiah Myers
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, United States
| | - Kymmia Petty
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, United States
| | - Rion Sam
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, United States
| | | | - Keshunna Reese
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, United States
| | - Linh Tran
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, United States
| | - Vaniyambadi Sridhar
- Department of Biology, University of New Orleans, New Orleans, LA, United States
| | - Christopher Williams
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, United States
| | - Jayalakshmi Sridhar
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, United States
| |
Collapse
|
6
|
Pan L, Li J, Xu Q, Gao Z, Yang M, Wu X, Li X. HER2/PI3K/AKT pathway in HER2-positive breast cancer: A review. Medicine (Baltimore) 2024; 103:e38508. [PMID: 38875362 PMCID: PMC11175886 DOI: 10.1097/md.0000000000038508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/01/2024] [Accepted: 05/17/2024] [Indexed: 06/16/2024] Open
Abstract
Breast cancer is currently the most commonly occurring cancer globally. Among breast cancer cases, the human epidermal growth factor receptor 2 (HER2)-positive breast cancer accounts for 15% to 20% and is a crucial focus in the treatment of breast cancer. Common HER2-targeted drugs approved for treating early and/or advanced breast cancer include trastuzumab and pertuzumab, which effectively improve patient prognosis. However, despite treatment, most patients with terminal HER2-positive breast cancer ultimately suffer death from the disease due to primary or acquired drug resistance. The prevalence of aberrantly activated the protein kinase B (AKT) signaling in HER2-positive breast cancer was already observed in previous studies. It is well known that p-AKT expression is linked to an unfavorable prognosis, and the phosphatidylinositol-3-kinase (PI3K)/AKT pathway, as the most common mutated pathway in breast cancer, plays a major role in the mechanism of drug resistance. Therefore, in the current review, we summarize the molecular alterations present in HER2-positive breast cancer, elucidate the relationships between HER2 overexpression and alterations in the PI3K/AKT signaling pathway and the pathways of the alterations in breast cancer, and summarize the resistant mechanism of drugs targeting the HER2-AKT pathway, which will provide an adjunctive therapeutic rationale for subsequent resistance to directed therapy in the future.
Collapse
Affiliation(s)
- Linghui Pan
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jinling Li
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Department of Laboratory Medicine, Chonggang General Hospital, Chongqing, China
| | - Qi Xu
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Zili Gao
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Mao Yang
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiaoping Wu
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xuesen Li
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
7
|
Yang L, Bhattacharya A, Peterson D, Li Y, Liu X, Marangoni E, Robila V, Zhang Y. Targeted dual degradation of HER2 and EGFR obliterates oncogenic signaling, overcomes therapy resistance, and inhibits metastatic lesions in HER2-positive breast cancer models. Drug Resist Updat 2024; 74:101078. [PMID: 38503142 PMCID: PMC11070302 DOI: 10.1016/j.drup.2024.101078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 03/21/2024]
Abstract
AIMS Human epidermal growth factor receptor 2 (HER2) is an oncogenic receptor tyrosine kinase amplified in approximately 20% of breast cancer (BC). HER2-targeted therapies are the linchpin of treating HER2-positive BC. However, drug resistance is common, and the main resistance mechanism is unknown. We tested the hypothesis that drug resistance results mainly from inadequate or lack of inhibition of HER2 and its family member epidermal growth factor receptor (EGFR). METHODS We used clinically relevant cell and tumor models to assess the impact of targeted degradation of HER2 and EGFR on trastuzumab resistance. Trastuzumab is the most common clinically used HER2 inhibitor. Targeted degradation of HER2 and EGFR was achieved using recombinant human protein PEPDG278D, which binds to the extracellular domains of the receptors. siRNA knockdown was used to assess the relative importance of EGFR and HER2 in trastuzumab resistance. RESULTS Both HER2 and EGFR are overexpressed in all trastuzumab-resistant HER2-positive BC cell and tumor models and that all trastuzumab-resistant models are highly vulnerable to targeted degradation of HER2 and EGFR. Degradation of HER2 and EGFR induced by PEPDG278D causes extensive inhibition of oncogenic signaling in trastuzumab-resistant HER2-positive BC cells. This is accompanied by strong growth inhibition of cultured cells, orthotopic patient-derived xenografts, and metastatic lesions in the brain and lung of trastuzumab-resistant HER2-positive BC. siRNA knockdown indicates that eliminating both HER2 and EGFR is necessary to maximize therapeutic outcome. CONCLUSIONS This study unravels the therapeutic vulnerability of trastuzumab-resistant HER2-positive BC and shows that an agent that targets the degradation of both HER2 and EGFR is highly effective in overcoming drug resistance in this disease. The findings provide new insights and innovations for advancing treatment of drug-resistant HER2-positive breast cancer that remains an unmet problem.
Collapse
Affiliation(s)
- Lu Yang
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Arup Bhattacharya
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Darrell Peterson
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, School of Pharmacy, Richmond, VA, USA
| | - Yun Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Xiaozhuo Liu
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Valentina Robila
- Department of Pathology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Yuesheng Zhang
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
8
|
Vo TH, EL-Sherbieny Abdelaal E, Jordan E, O'Donovan O, McNeela EA, Mehta JP, Rani S. miRNAs as biomarkers of therapeutic response to HER2-targeted treatment in breast cancer: A systematic review. Biochem Biophys Rep 2024; 37:101588. [PMID: 38088952 PMCID: PMC10711031 DOI: 10.1016/j.bbrep.2023.101588] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/19/2023] [Indexed: 06/16/2024] Open
Abstract
Breast cancer is the most common type of lethal cancer in women globally. Women have a 1 in 8 chance of developing breast cancer in their lifetime. Among the four primary molecular subtypes (luminal A, luminal B, HER2+, and triple-negative), HER2+ accounts for 20-25 % of all breast cancer and is rather aggressive. Although the treatment outcome of HER2+ breast cancer patients has been significantly improved with anti-HER2 agents, primary and acquired drug resistance present substantial clinical issues, limiting the benefits of HER2-targeted treatment. MicroRNAs (miRNAs) play a central role in regulating acquired drug resistance. miRNA are single-stranded, non-coding RNAs of around 20-25 nucleotides, known for essential roles in regulating gene expression at the post-transcriptional level. Increasing evidence has demonstrated that miRNA-mediated alteration of gene expression is associated with tumorigenesis, metastasis, and tumor response to treatment. Comprehensive knowledge of miRNAs as potential markers of drug response can help provide valuable guidance for treatment prognosis and personalized medicine for breast cancer patients.
Collapse
Affiliation(s)
- Thanh Hoa Vo
- Department of Science, School of Science and Computing, South East Technological University, Cork Road, Waterford, X91 K0EK, Ireland
- Pharmaceutical and Molecular Biotechnology Research Center, South East Technological University, Cork Road, X91 K0EK, Waterford, Ireland
| | | | - Emmet Jordan
- Department of Oncology, University Hospital Waterford, Dunmore Road, X91 ER8E, Waterford, Ireland
| | - Orla O'Donovan
- Department of Science, School of Science and Computing, South East Technological University, Cork Road, Waterford, X91 K0EK, Ireland
- Pharmaceutical and Molecular Biotechnology Research Center, South East Technological University, Cork Road, X91 K0EK, Waterford, Ireland
| | - Edel A. McNeela
- Department of Science, School of Science and Computing, South East Technological University, Cork Road, Waterford, X91 K0EK, Ireland
- Pharmaceutical and Molecular Biotechnology Research Center, South East Technological University, Cork Road, X91 K0EK, Waterford, Ireland
| | - Jai Prakash Mehta
- Department of Applied Science, South East Technological University, Kilkenny Road, R93 V960, Carlow, Ireland
| | - Sweta Rani
- Department of Science, School of Science and Computing, South East Technological University, Cork Road, Waterford, X91 K0EK, Ireland
- Pharmaceutical and Molecular Biotechnology Research Center, South East Technological University, Cork Road, X91 K0EK, Waterford, Ireland
| |
Collapse
|
9
|
Sanz-Álvarez M, Luque M, Morales-Gallego M, Cristóbal I, Ramírez-Merino N, Rangel Y, Izarzugaza Y, Eroles P, Albanell J, Madoz-Gúrpide J, Rojo F. Generation and Characterization of Trastuzumab/Pertuzumab-Resistant HER2-Positive Breast Cancer Cell Lines. Int J Mol Sci 2023; 25:207. [PMID: 38203378 PMCID: PMC10779249 DOI: 10.3390/ijms25010207] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
The combination of trastuzumab and pertuzumab as first-line therapy in patients with HER2-positive breast cancer has shown significant clinical benefits compared to trastuzumab alone. However, despite initial therapeutic success, most patients eventually progress, and tumors develop acquired resistance and invariably relapse. Therefore, there is an urgent need to improve our understanding of the mechanisms governing resistance in order to develop targeted therapeutic strategies with improved efficacy. We generated four novel HER2-positive cell lines via prolonged exposure to trastuzumab and pertuzumab and determined their resistance rates. Long-term resistance was confirmed by a significant increase in the colony-forming capacity of the derived cells. We authenticated the molecular identity of the new lines via both immunohistochemistry for the clinical phenotype and molecular profiling of point mutations. HER2 overexpression was confirmed in all resistant cell lines, and acquisition of resistance to trastuzumab and pertuzumab did not translate into differences in ER, PR, and HER2 receptor expression. In contrast, changes in the expression and activity of other HER family members, particularly HER4, were observed. In the same vein, analyses of the receptor and effector kinase status of different cellular pathways revealed that the MAPK pathway may be involved in the acquisition of resistance to trastuzumab and pertuzumab. Finally, proteomic analysis confirmed a significant change in the abundance patterns of more than 600 proteins with implications in key biological processes, such as ribosome formation, mitochondrial activity, and metabolism, which could be relevant mechanisms in the generation of resistance in HER2-positive breast cancer. We concluded that these resistant BCCLs may be a valuable tool to better understand the mechanisms of acquisition of resistance to trastuzumab and pertuzumab-based anti-HER2 therapy.
Collapse
Affiliation(s)
- Marta Sanz-Álvarez
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (M.L.); (M.M.-G.)
| | - Melani Luque
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (M.L.); (M.M.-G.)
| | - Miriam Morales-Gallego
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (M.L.); (M.M.-G.)
| | - Ion Cristóbal
- Translational Oncology Division, OncoHealth Institute, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain;
| | | | - Yamileth Rangel
- Department of Pathology, Infanta Elena University Hospital, 28342 Madrid, Spain;
| | - Yann Izarzugaza
- Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, 28040 Madrid, Spain;
| | - Pilar Eroles
- Institute of Health Research INCLIVA—CIBERONC, 46010 Valencia, Spain;
- Department of Physiology, University of Valencia, 46010 Valencia, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), 08003 Barcelona, Spain;
- Department of Medical Oncology, Hospital del Mar—CIBERONC, 08003 Barcelona, Spain
- Department of Experimental and Health Sciences, Faculty of Medicine, Universitat Pompeu Fabra, 08002 Barcelona, Spain
| | - Juan Madoz-Gúrpide
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (M.L.); (M.M.-G.)
| | - Federico Rojo
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (M.L.); (M.M.-G.)
| |
Collapse
|
10
|
Adam-Artigues A, Arenas EJ, Arribas J, Prat A, Cejalvo JM. AXL - a new player in resistance to HER2 blockade. Cancer Treat Rev 2023; 121:102639. [PMID: 37864955 DOI: 10.1016/j.ctrv.2023.102639] [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: 07/26/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/23/2023]
Abstract
HER2 is a driver in solid tumors, mainly breast, oesophageal and gastric cancer, through activation of oncogenic signaling pathways such as PI3K or MAPK. HER2 overexpression associates with aggressive disease and poor prognosis. Despite targeted anti-HER2 therapy has improved outcomes and is the current standard of care, resistance emerge in some patients, requiring additional therapeutic strategies. Several mechanisms, including the upregulation of receptors tyrosine kinases such as AXL, are involved in resistance. AXL signaling leads to cancer cell proliferation, survival, migration, invasion and angiogenesis and correlates with poor prognosis. In addition, AXL overexpression accompanied by a mesenchymal phenotype result in resistance to chemotherapy and targeted therapies. Preclinical studies show that AXL drives anti-HER2 resistance and metastasis through dimerization with HER2 and activation of downstream pathways in breast cancer. Moreover, AXL inhibition restores response to HER2 blockade in vitro and in vivo. Limited data in gastric and oesophageal cancer also support these evidences. Furthermore, AXL shows a strong value as a prognostic and predictive biomarker in HER2+ breast cancer patients, adding a remarkable translational relevance. Therefore, current studies enforce the potential of co-targeting AXL and HER2 to overcome resistance and supports the use of AXL inhibitors in the clinic.
Collapse
Affiliation(s)
| | - Enrique J Arenas
- Josep Carreras Leukaemia Research Institute, Spain; Center for Biomedical Network Research on Cancer (CIBERONC), Spain.
| | - Joaquín Arribas
- Center for Biomedical Network Research on Cancer (CIBERONC), Spain; Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO), Spain; Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Spain; Department of Biochemistry and Molecular Biology, Universitat Autónoma de Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Spain.
| | - Aleix Prat
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Spain; Department of Medical Oncology, Hospital Clínic de Barcelona, Spain; SOLTI Breast Cancer Research Group, Spain.
| | - Juan Miguel Cejalvo
- INCLIVA Biomedical Research Institute, Spain; Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO), Spain; Department of Medical Oncology, Hospital Clínico Universitario de València, Spain.
| |
Collapse
|
11
|
Zagami P, Boscolo Bielo L, Nicolò E, Curigliano G. HER2-positive breast cancer: cotargeting to overcome treatment resistance. Curr Opin Oncol 2023; 35:461-471. [PMID: 37621172 DOI: 10.1097/cco.0000000000000971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
PURPOSE OF REVIEW The introduction in clinical practice of anti-HER2 agents changed the prognosis of patients with HER2-positive (HER2+) breast cancer in both metastatic and early setting. Although the incomparable results obtained in the last years with the approval of new drugs targeting HER2, not all patients derive benefit from these treatments, experiencing primary or secondary resistance. The aim of this article is to review the data about cotargeting HER2 with different pathways (or epitopes of receptors) involved in its oncogenic signaling, as a mechanism to overcome resistance to anti-HER2 agents. RECENT FINDINGS Concordantly to the knowledge of the HER2+ breast cancer heterogeneity as well as new drugs, novel predictive biomarkers of response to anti-HER2 treatments are always raised helping to define target to overcome resistance. Cotargeting HER2 and hormone receptors is the most well known mechanism to improve benefit in HER2+/HR+ breast cancer. Additional HER2-cotargeting, such as, with PI3K pathway, as well as different HERs receptors or immune-checkpoints revealed promising results. SUMMARY HER2+ breast cancer is an heterogenous disease. Cotargeting HER2 with other signaling pathways involved in its mechanism of resistance may improve patient outcomes. Research efforts will continue to investigate novel targets and combinations to create more effective treatment regimes.
Collapse
Affiliation(s)
- Paola Zagami
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Lineberger comprehensive cancer center, University of North Carolina, Chapel hill, North Carolina
| | - Luca Boscolo Bielo
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Eleonora Nicolò
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
12
|
Kim Y, Bae YJ, Kim JH, Kim H, Shin SJ, Jung DH, Park H. Wnt/β-catenin pathway is a key signaling pathway to trastuzumab resistance in gastric cancer cells. BMC Cancer 2023; 23:922. [PMID: 37773114 PMCID: PMC10542239 DOI: 10.1186/s12885-023-11447-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/25/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Trastuzumab is the only approved target agent for the first-line treatment of human epidermal growth factor receptor-2 (HER-2) positive gastric cancer; however, trastuzumab resistance is a major problem in clinical practice. To comprehend the mechanism of trastuzumab resistance, we focused on the Wnt/β-catenin signaling pathway and its influence on the phenotypes and behavior of trastuzumab-resistant gastric cancer cells. METHODS Trastuzumab-resistant NCI-N87R cells were established in vitro from the human gastric cancer cell line NCI-N87 by dose-escalating repeated trastuzumab treatment. We investigated the phenotypes of NCI-N87R cells, including Wnt signaling pathway activity. Gastric cancer organoid cells were incubated with complete medium and Wnt3a-depletion medium, and their resistance to trastuzumab was compared. RESULTS NCI-N87R exhibited stemness and epithelial-mesenchymal transition (EMT)-like phenotypes, along with decreased levels of the epithelial marker E-cadherin and increased levels of the mesenchymal markers Vimentin and Snail along with an increased Wnt signaling pathway activity. When gastric cancer cells were incubated in Wnt3a-conditioned medium. Wnt signaling pathway activity and resistance to trastuzumab increased. Gastric cancer patient-derived organoids incubated in Wnt3a-depletion medium were more susceptible to dose-dependent inhibition of cell viability by trastuzumab than those incubated in complete medium. CONCLUSIONS Trastuzumab-resistant gastric cancer cells exhibited EMT-like phenotype, and trastuzumab resistance was promoted by the Wnt/β-catenin signaling pathway. The Wnt/β-catenin pathway is a key signaling pathway for trastuzumab resistance in gastric cancer cells.
Collapse
Affiliation(s)
- Yuna Kim
- Department of Internal Medicine, Division of Gastroenterology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20, Eonju-ro 63-gil, Gangnam-gu, Seoul, 06229, Korea
| | - Yoo Jin Bae
- Department of Internal Medicine, Division of Gastroenterology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20, Eonju-ro 63-gil, Gangnam-gu, Seoul, 06229, Korea
| | - Jie-Hyun Kim
- Department of Internal Medicine, Division of Gastroenterology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20, Eonju-ro 63-gil, Gangnam-gu, Seoul, 06229, Korea.
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Su-Jin Shin
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Korea
| | - Da Hyun Jung
- Department of Internal Medicine, Division of Gastroenterology, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hyojin Park
- Department of Internal Medicine, Division of Gastroenterology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20, Eonju-ro 63-gil, Gangnam-gu, Seoul, 06229, Korea
| |
Collapse
|
13
|
Wu X, Huang S, He W, Song M. Emerging insights into mechanisms of trastuzumab resistance in HER2-positive cancers. Int Immunopharmacol 2023; 122:110602. [PMID: 37437432 DOI: 10.1016/j.intimp.2023.110602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/19/2023] [Accepted: 07/02/2023] [Indexed: 07/14/2023]
Abstract
HER2 is an established therapeutic target in breast, gastric, and gastroesophageal junction carcinomas with HER2 overexpression or genomic alterations. The humanized monoclonal antibody trastuzumab targeting HER2 has substantially improved the clinical outcomes of HER2-positive patients, yet the inevitable intrinsic or acquired resistance to trastuzumab limits its clinical benefit, necessitating the elucidation of resistance mechanisms to develop alternate therapeutic strategies. This review presents an overview of trastuzumab resistance mechanisms involving signaling pathways, cellular metabolism, cell plasticity, and tumor microenvironment, particularly discussing the prospects of developing rational combinations to improve patient outcomes.
Collapse
Affiliation(s)
- Xiaoxue Wu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Shuting Huang
- School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Weiling He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China.
| | - Mei Song
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
14
|
Cao J, Teng Y, Li H, Zhang L, Ouyang Q, Xie W, Pan Y, Song Z, Ling X, Wu X, Xu J, Li L, Ren L, Wang H, Zhou D, Luo J, Hu X. Pyrotinib plus capecitabine for trastuzumab-resistant, HER2-positive advanced breast cancer (PICTURE): a single-arm, multicenter phase 2 trial. BMC Med 2023; 21:300. [PMID: 37559142 PMCID: PMC10410905 DOI: 10.1186/s12916-023-02999-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/23/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Patients with human epidermal growth factor receptor 2 (HER2)-positive advanced breast cancer and primary resistance to trastuzumab have a poor clinical outcome and lack good evidence to inform clinical decision. This study investigated the efficacy and safety of pyrotinib plus capecitabine in this population. METHODS This phase 2 trial was conducted at 16 sites in China. Patients received oral pyrotinib 400 mg once daily and capecitabine 1000 mg/m2 twice a day on days 1-14 of each 21-day cycle until disease progression or intolerable toxicity. The primary endpoint was investigator-assessed progression-free survival (PFS). RESULTS Between June 2019 and September 2021, 100 patients were enrolled with a median age of 51 years (range, 24-69). All patients had been treated with trastuzumab and 21 (21.0%) patients had prior use of pertuzumab. As of August 31, 2022, the median follow-up duration was 20.1 months (range, 1.3-38.2). The median PFS was 11.8 months (95% confidence interval [CI], 8.4-15.1), which crossed the pre-specified efficacy boundary of 8.0 months. The objective response rate was 70.0% (70/100), with a median duration of response of 13.8 months (95% CI, 10.2-19.3). The disease control rate was 87.0% (87/100). The median overall survival was not reached. The most common grade ≥ 3 treatment-emergent adverse event was diarrhea (24 [24.0%]). No treatment-related deaths occurred. CONCLUSIONS Pyrotinib plus capecitabine can be considered to be a treatment option in HER2-positive advanced breast cancer patients who have shown primary resistance to trastuzumab. Even in the era of modern anti-HER2 treatments, this clinical setting warrants more investigations to meet unmet needs. TRIAL REGISTRATION ClinicalTrials.gov, NCT04001621. Retrospectively registered on June 28, 2019.
Collapse
Affiliation(s)
- Jun Cao
- Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, China
| | - Yuee Teng
- Department of Medical Oncology, The First Hospital of China Medical University, 110001, Shenyang, China
| | - Huiping Li
- Department of Medical Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Lili Zhang
- Department of Medical Oncology, Jiangsu Cancer Hospital, Nanjing, 210008, China
| | - Quchang Ouyang
- Department of Medical Oncology, Hunan Cancer Hospital, Changsha, 410013, China
| | - Weimin Xie
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, 530027, China
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Zhenchuan Song
- Breast Center, The Fourth Hospital of Hebei Medical University, 050011, Shijiazhuang, China
| | - Xiaoling Ling
- Department of Medical Oncology, The First Hospital of Lanzhou University, Lanzhou, 730013, China
| | - Xiaohong Wu
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Jingwei Xu
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Li Li
- Department of Medical Oncology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Liping Ren
- Department of Breast Surgery, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Hong Wang
- Department of Medical Oncology, The Third Hospital of Nanchang, Nanchang, 330008, China
| | - Dongxian Zhou
- Department of Breast Surgery, Shenzhen People's Hospital, Shenzhen, 518020, China
| | - Jing Luo
- Department of Breast Surgery, Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Xichun Hu
- Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, China.
| |
Collapse
|
15
|
Liu H, Ruan S, Larsen ME, Tan C, Liu B, Lyu H. Trastuzumab-resistant breast cancer cells-derived tumor xenograft models exhibit distinct sensitivity to lapatinib treatment in vivo. Biol Proced Online 2023; 25:19. [PMID: 37370010 DOI: 10.1186/s12575-023-00212-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Resistance to HER2-targeted therapies, including the monoclonal antibody trastuzumab and tyrosine kinase inhibitor lapatinib, frequently occurs and currently represents a significant clinical challenge in the management of HER2-positive breast cancer. We previously showed that the trastuzumab-resistant SKBR3-pool2 and BT474-HR20 sublines were refractory to lapatinib in vitro as compared to the parental SKBR3 and BT474 cells, respectively. The in vivo efficacy of lapatinib against trastuzumab-resistant breast cancer remained unclear. RESULTS In tumor xenograft models, both SKBR3-pool2- and BT474-HR20-derived tumors retained their resistance phenotype to trastuzumab; however, those tumors responded differently to the treatment with lapatinib. While lapatinib markedly suppressed growth of SKBR3-pool2-derived tumors, it slightly attenuated BT474-HR20 tumor growth. Immunohistochemistry analyses revealed that lapatinib neither affected the expression of HER3, nor altered the levels of phosphorylated HER3 and FOXO3a in vivo. Interestingly, lapatinib treatment significantly increased the levels of phosphorylated Akt and upregulated the expression of insulin receptor substrate-1 (IRS1) in the tumors-derived from BT474-HR20, but not SKBR3-pool2 cells. CONCLUSIONS Our data indicated that SKBR3-pool2-derived tumors were highly sensitive to lapatinib treatment, whereas BT474-HR20 tumors exhibited resistance to lapatinib. It seemed that the inefficacy of lapatinib against BT474-HR20 tumors in vivo was attributed to lapatinib-induced upregulation of IRS1 and activation of Akt. Thus, the tumor xenograft models-derived from SKBR3-pool2 and BT474-HR20 cells serve as an excellent in vivo system to test the efficacy of other HER2-targeted therapies and novel agents to overcome trastuzumab resistance against HER2-positive breast cancer.
Collapse
Affiliation(s)
- Hao Liu
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Sanbao Ruan
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA
| | - Margaret E Larsen
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA
| | - Congcong Tan
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA
| | - Bolin Liu
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA.
| | - Hui Lyu
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA.
| |
Collapse
|
16
|
Liu S, Xie SM, Liu W, Gagea M, Hanker AB, Nguyen N, Singareeka Raghavendra A, Yang-Kolodji G, Chu F, Neelapu SS, Marchese A, Hanash S, Zimmermann J, Arteaga CL, Tripathy D. Targeting CXCR4 abrogates resistance to trastuzumab by blocking cell cycle progression and synergizes with docetaxel in breast cancer treatment. Breast Cancer Res 2023; 25:62. [PMID: 37280713 DOI: 10.1186/s13058-023-01665-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 05/25/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Although trastuzumab and other HER2-targeted therapies have significantly improved survival in patients with HER2 overexpressed or amplified (HER2+) breast cancer, a significant proportion of patients do not respond or eventually develop clinical resistance. Strategies to reverse trastuzumab resistance remain a high clinical priority. We were the first to report the role of CXCR4 in trastuzumab resistance. The present study aims to explore the therapeutic potential of targeting CXCR4 and better understand the associated mechanisms. METHODS Immunofluorescent staining, confocal microscopy analysis, and immunoblotting were used to analyze CXCR4 expression. BrdU incorporation assays and flow cytometry were used to analyze dynamic CXCR4 expression. Three-dimensional co-culture (tumor cells/breast cancer-associated fibroblasts/human peripheral blood mononuclear cells) or antibody-dependent cellular cytotoxicity assay was used to mimic human tumor microenvironment, which is necessary for testing therapeutic effects of CXCR4 inhibitor or trastuzumab. The FDA-approved CXCR4 antagonist AMD3100, trastuzumab, and docetaxel chemotherapy were used to evaluate therapeutic efficacy in vitro and in vivo. Reverse phase protein array and immunoblotting were used to discern the associated molecular mechanisms. RESULTS Using a panel of cell lines and patient breast cancer samples, we confirmed CXCR4 drives trastuzumab resistance in HER2+ breast cancer and further demonstrated the increased CXCR4 expression in trastuzumab-resistant cells is associated with cell cycle progression with a peak in the G2/M phases. Blocking CXCR4 with AMD3100 inhibits cell proliferation by downregulating mediators of G2-M transition, leading to G2/M arrest and abnormal mitosis. Using a panel of trastuzumab-resistant cell lines and an in vivo established trastuzumab-resistant xenograft mouse model, we demonstrated that targeting CXCR4 with AMD3100 suppresses tumor growth in vitro and in vivo, and synergizes with docetaxel. CONCLUSIONS Our findings support CXCR4 as a novel therapeutic target and a predictive biomarker for trastuzumab resistance in HER2+ breast cancer.
Collapse
Affiliation(s)
- Shuying Liu
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shelly M Xie
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wenbin Liu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mihai Gagea
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ariella B Hanker
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nguyen Nguyen
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Gloria Yang-Kolodji
- Department of Medicine, University of South California, Los Angeles, CA, USA
| | - Fuliang Chu
- Department of Lymphoma-Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sattva S Neelapu
- Department of Lymphoma-Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Carlos L Arteaga
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Debasish Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
17
|
Yin L, Chen GL, Xiang Z, Liu YL, Li XY, Bi JW, Wang Q. Current progress in chimeric antigen receptor-modified T cells for the treatment of metastatic breast cancer. Biomed Pharmacother 2023; 162:114648. [PMID: 37023621 DOI: 10.1016/j.biopha.2023.114648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Breast cancer is the leading cancer in women. Around 20-30% breast cancer patients undergo invasion or metastasis after radical surgical resection and eventually die. Number of breast cancer patients show poor sensitivity toward treatments despite the advances in chemotherapy, endocrine therapy, and molecular targeted treatments. Therapeutic resistance and tumor recurrence or metastasis develop with the ongoing treatments. Conducive treatment strategies are thus required. Chimeric antigen receptor (CAR)-modified T-cell therapy has progressed as a part of tumor immunotherapy. However, CAR-T treatment has not been effective in solid tumors because of tumor microenvironment complexity, inhibitory effects of extracellular matrix, and lacking ideal tumor antigens. Herein, the prospects of CAR-T cell therapy for metastatic breast cancer are discussed, and the targets for CAR-T therapy in breast cancer (HER-2, C-MET, MSLN, CEA, MUC1, ROR1, EGFR) at clinical level are reviewed. Moreover, solutions are proposed for the challenges of breast cancer CAR-T therapy regarding off-target effects, heterogeneous antigen expression by tumor cells and immunosuppressive tumor microenvironment. Ideas for improving the therapeutics of CAR-T cell therapy in metastatic breast cancer are suggested.
Collapse
Affiliation(s)
- Li Yin
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China; Shandong University of Traditional Chinese Medicine, 250355 Jinan, China
| | - Gui-Lai Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China
| | - Zhuo Xiang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China
| | - Yu-Lin Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China
| | - Xing-Yu Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003 Qingdao, China
| | - Jing-Wang Bi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China.
| | - Qiang Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China; Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003 Qingdao, China.
| |
Collapse
|
18
|
He M, Chen ZF, Zhang L, Gao X, Chong X, Li HS, Shen L, Ji J, Zhang X, Dong B, Li ZY, Lei T. Associations of subcutaneous fat area and Systemic Immune-inflammation Index with survival in patients with advanced gastric cancer receiving dual PD-1 and HER2 blockade. J Immunother Cancer 2023; 11:e007054. [PMID: 37349127 PMCID: PMC10314655 DOI: 10.1136/jitc-2023-007054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Systemic Immune-inflammation Index (SII) and body composition parameters are easily assessed, and can predict overall survival (OS) in various cancers, allowing early intervention. This study aimed to assess the correlation between CT-derived body composition parameters and SII and OS in patients with advanced gastric cancer receiving dual programmed death-1 (PD-1) and human epidermal growth factor receptor 2 (HER2) blockade. MATERIALS AND METHODS This retrospective study enrolled patients with advanced gastric cancer treated with dual PD-1 and HER2 blockade from March 2019 to June 2022. We developed a deep learning model based on nnU-Net to automatically segment skeletal muscle, subcutaneous fat and visceral fat at the third lumbar level, and calculated the corresponding Skeletal Muscle Index, skeletal muscle density, subcutaneous fat area (SFA) and visceral fat area. SII was computed using the formula that total peripheral platelet count×neutrophil/lymphocyte ratio. Univariate and multivariate Cox regression analysis were used to determine the associations between SII, body composition parameters and OS. RESULTS The automatic segmentation deep learning model was developed to efficiently segment body composition in 158 patients (0.23 s/image). Multivariate Cox analysis revealed that high SII (HR=2.49 (95% CI 1.54 to 4.01), p<0.001) and high SFA (HR=0.42 (95% CI 0.24 to 0.73), p=0.002) were independently associated with OS, whereas sarcopenia was not an independent prognostic factor for OS (HR=1.41 (95% CI 0.86 to 2.31), p=0.173). In further analysis, patients with high SII and low SFA had worse long-term prognosis compared with those with low SII and high SFA (HR=8.19 (95% CI 3.91 to 17.16), p<0.001). CONCLUSION Pretreatment SFA and SII were significantly associated with OS in patients with advanced gastric cancer. A comprehensive analysis of SII and SFA may improve the prognostic stratification of patients with gastric cancer receiving dual PD-1 and HER2 blockade.
Collapse
Affiliation(s)
- Meng He
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
| | - Zi-Fan Chen
- Center for Data Science, Peking University, Beijing, China
| | - Li Zhang
- Center for Data Science, Peking University, Beijing, China
| | - Xiangyu Gao
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
| | - Xiaoyi Chong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Hao-Shen Li
- Center for Data Science, Peking University, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiafu Ji
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Bin Dong
- Beijing International Center for Mathematical Research, Peking University; Center for Machine Learning Research, Peking University; National Biomedical Imaging Center, Peking University, Beijing, China
| | - Zi-Yu Li
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
| | - Tang Lei
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China
| |
Collapse
|
19
|
Zhang XN, Gao Y, Zhang XY, Guo NJ, Hou WQ, Wang SW, Zheng YC, Wang N, Liu HM, Wang B. Detailed curriculum vitae of HER2-targeted therapy. Pharmacol Ther 2023; 245:108417. [PMID: 37075933 DOI: 10.1016/j.pharmthera.2023.108417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
With the booming development of precision medicine, molecular targeted therapy has been widely used in clinical oncology treatment due to a smaller number of side effects and its superior accuracy compared to that of traditional strategies. Among them, human epidermal growth factor receptor 2 (HER2)-targeted therapy has attracted considerable attention and has been used in the clinical treatment of breast and gastric cancer. Despite excellent clinical effects, HER2-targeted therapy remains in its infancy due to its resulting inherent and acquired resistance. Here, a comprehensive overview of HER2 in numerous cancers is presented, including its biological role, involved signaling pathways, and the status of HER2-targeted therapy.
Collapse
Affiliation(s)
- Xiao-Nan Zhang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Xi-Ya Zhang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Ning-Jie Guo
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Wen-Qing Hou
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Shu-Wu Wang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Ning Wang
- The School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China.
| | - Bo Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
20
|
Bashraheel SS, Kheraldine H, Khalaf S, Moustafa AEA. Metformin and HER2-positive breast cancer: Mechanisms and therapeutic implications. Biomed Pharmacother 2023; 162:114676. [PMID: 37037091 DOI: 10.1016/j.biopha.2023.114676] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/01/2023] [Accepted: 04/06/2023] [Indexed: 04/12/2023] Open
Abstract
Due to the strong association between diabetes and cancer incidents, several anti-diabetic drugs, including metformin, have been examined for their anticancer activity. Metformin is a biguanide antihyperglycemic agent used as a first-line drug for type II diabetes mellitus. It exhibits anticancer activity by impacting different molecular pathways, such as AMP-inducible protein kinase (AMPK)-dependent and AMPK-independent pathways. Additionally, Metformin indirectly inhibits IGF-1R signaling, which is highly activated in breast malignancy. On the other hand, breast cancer is one of the major causes of cancer-related morbidity and mortality worldwide, where the human epidermal growth factor receptor-positive (HER2-positive) subtype is one of the most aggressive ones with a high rate of lymph node metastasis. In this review, we summarize the association between diabetes and human cancer, listing recent evidence of metformin's anticancer activity. A special focus is dedicated to HER2-positive breast cancer with regards to the interaction between HER2 and IGF-1R. Then, we discuss combination therapy strategies of metformin and other anti-diabetic drugs in HER2-positive breast cancer.
Collapse
Affiliation(s)
| | - Hadeel Kheraldine
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Sarah Khalaf
- College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar; Biomedical Research Center, QU Health, Qatar University, PO. Box 2713, Doha, Qatar; Oncology Department, McGill University, Montreal, Quebec H3A 0G4, Canada.
| |
Collapse
|
21
|
Fang X, Xu J, Jin K, Qian J. Combining of immunotherapeutic approaches with chemotherapy for treatment of gastric cancer: Achievements and limitations. Int Immunopharmacol 2023; 118:110062. [PMID: 36965367 DOI: 10.1016/j.intimp.2023.110062] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/09/2023] [Accepted: 03/19/2023] [Indexed: 03/27/2023]
Abstract
Evidence reveals that gastric cancer (GC) is the fifth most common malignancy in humans, and about 770,000 people die from this cancer yearly. It has been reported that new cases and deaths from GC are more common in men than women. Therapeutic approaches, such as surgery, chemotherapy, and radiotherapy, have been common for treating GC. Nevertheless, due to the complications and limitations of these methods, researchers use novel approaches, such as immunotherapeutic or target therapies, to evaluate the effectiveness of treatment in patients with metastatic GC. Studies have shown that monotherapy is usually associated with unpromising outcomes, and combination therapy can be a more practical option for treating metastatic GC. Therefore, to clarify different aspects of chemotherapy and immunotherapy in patients with metastatic GC, this review discussed the achievements and challenges of combining immunotherapeutic methods with chemotherapeutic agents.
Collapse
Affiliation(s)
- Xingliang Fang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Jinfang Xu
- Department of Emergency Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, China
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People's Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang, Zhejiang 312500, China.
| |
Collapse
|
22
|
Liu S, Xie SM, Liu W, Gagea M, Hanker AB, Nguyen N, Raghavendra AS, Yang-Kolodji G, Chu F, Neelapu SS, Hanash S, Zimmermann J, Arteaga CL, Tripathy D. Targeting CXCR4 abrogates resistance to trastuzumab by blocking cell cycle progression and synergizes with docetaxel in breast cancer treatment. RESEARCH SQUARE 2023:rs.3.rs-2388864. [PMID: 36824840 PMCID: PMC9949251 DOI: 10.21203/rs.3.rs-2388864/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Background: Although trastuzumab and other HER2-targeted therapies have significantly improved survival in patients with HER2 overexpressed or amplified (HER2+) breast cancer, a significant proportion of patients do not respond or eventually develop clinical resistance. Strategies to reverse trastuzumab resistance remain a high clinical priority. We were the first to report the role of CXCR4 in trastuzumab resistance. The present study aims to explore the therapeutic potential of targeting CXCR4 and better understand the associated mechanisms. Methods: Immunofluorescent staining, confocal microscopy analysis, and immunoblotting were used to analyze CXCR4 expression. BrdU incorporation assays and flow cytometry were used to analyze dynamic CXCR4expression. Three-dimensional co-culture (tumor cells/ breast cancer-associated fibroblasts / human peripheral blood mononuclear cells) or antibody-dependent cellular cytotoxicity assay was used to mimic human tumor microenvironment, which is necessary for testing therapeutic effect of CXCR4 inhibitor or trastuzumab. The FDA-approved CXCR4 antagonist AMD3100, trastuzumab, and docetaxel chemotherapy were used to evaluate therapeutic efficacy in vitro and in vivo. Reverse phase protein array and immunoblotting were used to discern the associated molecular mechanisms. Results: Using multiple cell lines and patient breast cancer samples we confirmed CXCR4 drives trastuzumab resistance in HER2+ breast cancer and further demonstrated that the increased CXCR4 expression in trastuzumab-resistant cells is associated with cell cycle progression with a peak in the G2/M phases. Blocking CXCR4 with AMD3100 inhibits cell proliferation by downregulating mediators of G2-M transition, leading to G2/M arrest and abnormal mitosis. Using multiple trastuzumab-resistant cell lines and an in vivo established trastuzumab-resistant xenograft mouse model, we demonstrated that targeting CXCR4 with AMD3100 suppresses tumor growth in vitro and in vivo, and synergizes with docetaxel. Conclusions: Our findings support CXCR4 as a novel therapeutic target and a predictive biomarker for trastuzumab resistance in HER2+ breast cancer.
Collapse
Affiliation(s)
- Shuying Liu
- The University of Texas MD Anderson Cancer Center
| | | | - Wenbin Liu
- The University of Texas MD Anderson Cancer Center
| | - Mihai Gagea
- The University of Texas MD Anderson Cancer Center
| | | | | | | | | | - Fuliang Chu
- The University of Texas MD Anderson Cancer Center
| | | | - Samir Hanash
- The University of Texas MD Anderson Cancer Center
| | | | | | | |
Collapse
|
23
|
Normann LS, Haugen MH, Hongisto V, Aure MR, Leivonen SK, Kristensen VN, Tahiri A, Engebraaten O, Sahlberg KK, Mælandsmo GM. High-throughput screen in vitro identifies dasatinib as a candidate for combinatorial treatment with HER2-targeting drugs in breast cancer. PLoS One 2023; 18:e0280507. [PMID: 36706086 PMCID: PMC9882887 DOI: 10.1371/journal.pone.0280507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 01/02/2023] [Indexed: 01/28/2023] Open
Abstract
Human epidermal growth factor receptor 2-positive (HER2+) breast cancer is an aggressive subtype of this disease. Targeted treatment has improved outcome, but there is still a need for new therapeutic strategies as some patients respond poorly to treatment. Our aim was to identify compounds that substantially affect viability in HER2+ breast cancer cells in response to combinatorial treatment. We performed a high-throughput drug screen of 278 compounds in combination with trastuzumab and lapatinib using two HER2+ breast cancer cell lines (KPL4 and SUM190PT). The most promising drugs were validated in vitro and in vivo, and downstream molecular changes of the treatments were analyzed. The screen revealed multiple drugs that could be used in combination with lapatinib and/or trastuzumab. The Src-inhibitor dasatinib showed the largest combinatorial effect together with lapatinib in the KPL4 cell line compared to treatment with dasatinib alone (p < 0.01). In vivo, only lapatinib significantly reduced tumor growth (p < 0.05), whereas dasatinib alone, or in combination with lapatinib, did not show significant effects. Protein analyses of the treated xenografts showed significant alterations in protein levels compared to untreated controls, suggesting that all drugs reached the tumor and exerted a measurable effect. In silico analyses suggested activation of apoptosis and reduced activity of survival pathways by all treatments, but the opposite pattern was observed for the combinatorial treatment compared to lapatinib alone.
Collapse
Affiliation(s)
- Lisa Svartdal Normann
- Department of Research and Innovation, Vestre Viken Hospital Trust, Drammen, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Mads Haugland Haugen
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Vesa Hongisto
- Division of Toxicology, Misvik Biology, Turku, Finland
| | - Miriam Ragle Aure
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Suvi-Katri Leivonen
- Applied Tumor Genomics Research Program, Medical Faculty, University of Helsinki, Helsinki, Finland
| | - Vessela N. Kristensen
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Andliena Tahiri
- Department of Research and Innovation, Vestre Viken Hospital Trust, Drammen, Norway
| | - Olav Engebraaten
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kristine Kleivi Sahlberg
- Department of Research and Innovation, Vestre Viken Hospital Trust, Drammen, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- * E-mail:
| | - Gunhild Mari Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute for Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
24
|
Zhang X. Molecular Classification of Breast Cancer: Relevance and Challenges. Arch Pathol Lab Med 2023; 147:46-51. [PMID: 36136295 DOI: 10.5858/arpa.2022-0070-ra] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2022] [Indexed: 12/31/2022]
Abstract
CONTEXT.— Appropriate patient management requires precise and meaningful tumor classification. Breast cancer classification continues to evolve from traditional morphologic evaluation to more sophisticated systems with the integration of new knowledge from research being translated into practice. Breast cancer is heterogeneous at the molecular level, with diversified patterns of gene expression, which is presumably responsible for the difference in tumor behavior and prognosis. Since the beginning of this century, new molecular technology has been gradually applied to breast cancer research on issues pertinent to prognosis (prognostic signature) and therapeutic prediction (predictive signature), and much progress has been made. OBJECTIVE.— To summarize the current state and the prospective future of molecular classification of breast cancer. DATA SOURCES.— Sources include recent medical literature on molecular classification of breast cancer. CONCLUSIONS.— Identification of intrinsic tumor subtypes has set a foundation for refining the breast cancer molecular classification. Studies have explored the genetic features within the intrinsic cancer subtypes and have identified novel molecular targets that led to the innovation of clinical assays to predict a patient's prognosis and to provide specific guidelines for therapeutic decisions. With the development and implication of these molecular tools, we have remarkably advanced our knowledge and enhanced our power to provide optimal management to patients. However, challenges still exist. Besides accurate prediction of prognosis, we are still in urgent need of more molecular predictors for tumor response to therapeutic regimes. Further exploration along this path will be critical for improving a patient's prognosis.
Collapse
Affiliation(s)
- Xinmin Zhang
- From the Department of Pathology, Cooper University Hospital, Cooper Medical School of Rowan University, Camden, New Jersey
| |
Collapse
|
25
|
Negi A, Kesari KK, Voisin-Chiret AS. Estrogen Receptor-α Targeting: PROTACs, SNIPERs, Peptide-PROTACs, Antibody Conjugated PROTACs and SNIPERs. Pharmaceutics 2022; 14:pharmaceutics14112523. [PMID: 36432713 PMCID: PMC9699327 DOI: 10.3390/pharmaceutics14112523] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Targeting selective estrogen subtype receptors through typical medicinal chemistry approaches is based on occupancy-driven pharmacology. In occupancy-driven pharmacology, molecules are developed in order to inhibit the protein of interest (POI), and their popularity is based on their virtue of faster kinetics. However, such approaches have intrinsic flaws, such as pico-to-nanomolar range binding affinity and continuous dosage after a time interval for sustained inhibition of POI. These shortcomings were addressed by event-driven pharmacology-based approaches, which degrade the POI rather than inhibit it. One such example is PROTACs (Proteolysis targeting chimeras), which has become one of the highly successful strategies of event-driven pharmacology (pharmacology that does the degradation of POI and diminishes its functions). The selective targeting of estrogen receptor subtypes is always challenging for chemical biologists and medicinal chemists. Specifically, estrogen receptor α (ER-α) is expressed in nearly 70% of breast cancer and commonly overexpressed in ovarian, prostate, colon, and endometrial cancer. Therefore, conventional hormonal therapies are most prescribed to patients with ER + cancers. However, on prolonged use, resistance commonly developed against these therapies, which led to selective estrogen receptor degrader (SERD) becoming the first-line drug for metastatic ER + breast cancer. The SERD success shows that removing cellular ER-α is a promising approach to overcoming endocrine resistance. Depending on the mechanism of degradation of ER-α, various types of strategies of developed.
Collapse
Affiliation(s)
- Arvind Negi
- Department of Bioproduct and Biosystems, Aalto University, 00076 Espoo, Finland
- Correspondence: or (A.N.); or (K.K.K.); (A.S.V.-C.)
| | - Kavindra Kumar Kesari
- Department of Bioproduct and Biosystems, Aalto University, 00076 Espoo, Finland
- Department of Applied Physics, School of Science, Aalto University, 02150 Espoo, Finland
- Correspondence: or (A.N.); or (K.K.K.); (A.S.V.-C.)
| | - Anne Sophie Voisin-Chiret
- CERMN (Centre d’Etudes et de Recherche sur le Médicament de Normandie), Normandie University UNICAEN, 14000 Caen, France
- Correspondence: or (A.N.); or (K.K.K.); (A.S.V.-C.)
| |
Collapse
|
26
|
Najjar MK, Manore SG, Regua AT, Lo HW. Antibody-Drug Conjugates for the Treatment of HER2-Positive Breast Cancer. Genes (Basel) 2022; 13:2065. [PMID: 36360302 PMCID: PMC9691220 DOI: 10.3390/genes13112065] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) receptor tyrosine kinase is overexpressed in 20-30% of breast cancers and is associated with poor prognosis and worse overall patient survival. Most women with HER2-positive breast cancer receive neoadjuvant chemotherapy plus HER2-targeted therapies. The development of HER2-directed therapeutics is an important advancement in targeting invasive breast cancer. Despite the efficacy of anti-HER2 monoclonal antibodies, they are still being combined with adjuvant chemotherapy to improve overall patient outcomes. Recently, significant progress has been made towards the development of a class of therapeutics known as antibody-drug conjugates (ADCs), which leverage the high specificity of HER2-targeted monoclonal antibodies with the potent cytotoxic effects of various small molecules, such as tubulin inhibitors and topoisomerase inhibitors. To date, two HER2-targeting ADCs have been approved by the FDA for the treatment of HER2-positive breast cancer: Ado-trastuzumab emtansine (T-DM1; Kadcyla®) and fam-trastuzumab deruxtecan-nxki (T-Dxd; Enhertu®). Kadcyla and Enhertu are approved for use as a second-line treatment after trastuzumab-taxane-based therapy in patients with HER2-positive breast cancer. The success of ADCs in the treatment of HER2-positive breast cancer provides novel therapeutic advancements in the management of the disease. In this review, we discuss the basic biology of HER2, its downstream signaling pathways, currently available anti-HER2 therapeutic modalities and their mechanisms of action, and the latest clinical and safety characteristics of ADCs used for the treatment of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Mariana K. Najjar
- Wake Forest Graduate School of Biomedical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Sara G. Manore
- Wake Forest Graduate School of Biomedical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Angelina T. Regua
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, MSE R162, 6431 Fannin Street, Houston, TX 77030, USA
| | - Hui-Wen Lo
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, MSE R162, 6431 Fannin Street, Houston, TX 77030, USA
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
27
|
Resistance to Trastuzumab. Cancers (Basel) 2022; 14:cancers14205115. [PMID: 36291900 PMCID: PMC9600208 DOI: 10.3390/cancers14205115] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Trastuzumab is a humanized antibody that has significantly improved the management and treatment outcomes of patients with cancers that overexpress HER2. Many research groups, both in academia and industry, have contributed towards understanding the various mechanisms engaged by trastuzumab to mediate its anti-tumor effects. Nevertheless, data from several clinical studies have indicated that a significant proportion of patients exhibit primary or acquired resistance to trastuzumab therapy. In this article, we discuss underlying mechanisms that contribute towards to resistance. Furthermore, we discuss the potential strategies to overcome some of the mechanisms of resistance to enhance the therapeutic efficacy of trastuzumab and other therapies based on it. Abstract One of the most impactful biologics for the treatment of breast cancer is the humanized monoclonal antibody, trastuzumab, which specifically recognizes the HER2/neu (HER2) protein encoded by the ERBB2 gene. Useful for both advanced and early breast cancers, trastuzumab has multiple mechanisms of action. Classical mechanisms attributed to trastuzumab action include cell cycle arrest, induction of apoptosis, and antibody-dependent cell-mediated cytotoxicity (ADCC). Recent studies have identified the role of the adaptive immune system in the clinical actions of trastuzumab. Despite the multiple mechanisms of action, many patients demonstrate resistance, primary or adaptive. Newly identified molecular and cellular mechanisms of trastuzumab resistance include induction of immune suppression, vascular mimicry, generation of breast cancer stem cells, deregulation of long non-coding RNAs, and metabolic escape. These newly identified mechanisms of resistance are discussed in detail in this review, particularly considering how they may lead to the development of well-rationalized, patient-tailored combinations that improve patient survival.
Collapse
|
28
|
Gámez-Chiachio M, Sarrió D, Moreno-Bueno G. Novel Therapies and Strategies to Overcome Resistance to Anti-HER2-Targeted Drugs. Cancers (Basel) 2022; 14:4543. [PMID: 36139701 PMCID: PMC9496705 DOI: 10.3390/cancers14184543] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
The prognosis and quality of life of HER2 breast cancer patients have significantly improved due to the crucial clinical benefit of various anti-HER2 targeted therapies. However, HER2 tumors can possess or develop several resistance mechanisms to these treatments, thus leaving patients with a limited set of additional therapeutic options. Fortunately, to overcome this problem, in recent years, multiple different and complementary approaches have been developed (such as antibody-drug conjugates (ADCs)) that are in clinical or preclinical stages. In this review, we focus on emerging strategies other than on ADCs that are either aimed at directly target the HER2 receptor (i.e., novel tyrosine kinase inhibitors) or subsequent intracellular signaling (e.g., PI3K/AKT/mTOR, CDK4/6 inhibitors, etc.), as well as on innovative approaches designed to attack other potential tumor weaknesses (such as immunotherapy, autophagy blockade, or targeting of other genes within the HER2 amplicon). Moreover, relevant technical advances such as anti-HER2 nanotherapies and immunotoxins are also discussed. In brief, this review summarizes the impact of novel therapeutic approaches on current and future clinical management of aggressive HER2 breast tumors.
Collapse
Affiliation(s)
- Manuel Gámez-Chiachio
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - David Sarrió
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - Gema Moreno-Bueno
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
- MD Anderson International Foundation, 28033 Madrid, Spain
| |
Collapse
|
29
|
ACBD3 Bioinformatic Analysis and Protein Expression in Breast Cancer Cells. Int J Mol Sci 2022; 23:ijms23168881. [PMID: 36012147 PMCID: PMC9408326 DOI: 10.3390/ijms23168881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/28/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
ACBD3 overexpression has previously been found to correlate with worse prognosis for breast cancer patients and, as an incredibly diverse protein in both function and cellular localisation, ACBD3 may have a larger role in breast cancer than previously thought. This study further investigated ACBD3′s role in breast cancer. Bioinformatic databases were queried to characterise ACBD3 expression and mutation in breast cancer and to investigate how overexpression affects breast cancer patient outcomes. Immunohistochemistry was carried out to examine ACBD3 location within cells and tissue structures. ACBD3 was more highly expressed in breast cancer than in any other cancer or matched normal tissue, and expression over the median level resulted in reduced relapse-free, overall, and distant metastasis-free survival for breast cancer patients as a whole, with some differences observed between subtypes. IHC analysis found that ACBD3 levels varied based on hormone receptor status, indicating that ACBD3 could be a candidate biomarker for poor patient prognosis in breast cancer and may possibly be a biomarker for ER signal reprogramming of precancerous breast tissue.
Collapse
|
30
|
Sridhar J, Komati R, Kumar S. Targeting RPS6K1 for Refractory Breast Cancer Therapy. Breast Cancer 2022. [DOI: 10.36255/exon-publications-breast-cancer-rps6k1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
31
|
Liu Y, Yu S, Xu T, Bodenko V, Orlova A, Oroujeni M, Rinne SS, Tolmachev V, Vorobyeva A, Gräslund T. Preclinical Evaluation of a New Format of 68Ga- and 111In-Labeled Affibody Molecule Z IGF-1R:4551 for the Visualization of IGF-1R Expression in Malignant Tumors Using PET and SPECT. Pharmaceutics 2022; 14:pharmaceutics14071475. [PMID: 35890370 PMCID: PMC9320461 DOI: 10.3390/pharmaceutics14071475] [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: 06/22/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
The Insulin-like growth factor-1 receptor (IGF-1R) is a molecular target for several monoclonal antibodies undergoing clinical evaluation as anticancer therapeutics. The non-invasive detection of IGF-1R expression in tumors might enable stratification of patients for specific treatment and improve the outcome of both clinical trials and routine treatment. The affibody molecule ZIGF-1R:4551 binds specifically to IGF-1R with subnanomolar affinity. The goal of this study was to evaluate the 68Ga and 111In-labeled affibody construct NODAGA-(HE)3-ZIGF-1R:4551 for the imaging of IGF-1R expression, using PET and SPECT. The labeling was efficient and provided stable coupling of both radionuclides. The two imaging probes, [68Ga]Ga-NODAGA-(HE)3-ZIGF-1R:4551 and [111In]In-NODAGA-(HE)3-ZIGF-1R:4551, demonstrated specific binding to IGF-1R-expressing human cancer cell lines in vitro and to IGF-1R-expressing xenografts in mice. Preclinical PET and SPECT/CT imaging demonstrated visualization of IGF-1R-expressing xenografts already one hour after injection. The tumor-to-blood ratios at 3 h after injection were 7.8 ± 0.2 and 8.0 ± 0.6 for [68Ga]Ga-NODAGA-(HE)3-ZIGF-1R:4551 and [111In]In-NODAGA-(HE)3-ZIGF-1R:4551, respectively. In conclusion, a molecular design of the ZIGF-1R:4551 affibody molecule, including placement of a (HE)3-tag on the N-terminus and site-specific coupling of a NODAGA chelator on the C-terminus, provides a tracer with improved imaging properties for visualization of IGF-1R in malignant tumors, using PET and SPECT.
Collapse
Affiliation(s)
- Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Shengze Yu
- Department of Protein Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden;
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Vitalina Bodenko
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
- Department of Medicinal Chemistry, Uppsala University, 75123 Uppsala, Sweden;
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
- Affibody AB, 17165 Solna, Sweden
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 75123 Uppsala, Sweden;
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
- Correspondence: (V.T.); (T.G.); Tel.: +46-704-250782 (V.T.); +46-8790-9627 (T.G.)
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden;
- Correspondence: (V.T.); (T.G.); Tel.: +46-704-250782 (V.T.); +46-8790-9627 (T.G.)
| |
Collapse
|
32
|
Li X, Sun B, Zhu J, Qian M, Chen Y. Construction of a Mass-Tagged Oligo Probe Set for Revealing Protein Ratiometric Relationship Associated with EGFR-HER2 Heterodimerization in Living Cells. Anal Chem 2022; 94:8838-8846. [PMID: 35709389 DOI: 10.1021/acs.analchem.1c04989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein dimerization, as the most common form of protein-protein interaction, can manifest more significant roles in cellular signaling than individual monomers. For example, excessive formation of EGFR-HER2 dimer has been implicated in cancer development and therapeutic resistance in addition to the overexpression of EGFR and HER2 proteins. Thus, quantitative evaluation of these heterodimers in living cells and revelation of their ratiometric relationship with protein monomers in dimerization may provide insights into clinical cancer management. To achieve this goal, the prerequisite is protein heterodimer quantification. Given the current lack of quantitative methods, we constructed a mass-tagged oligo nanoprobe set for quantification of EGFR-HER2 dimer in living cells. The mass-tagged oligo nanoprobe set contained two targeting probes (nucleic acid aptamers), a connector probe, a hairpin probe, and a photocleavable mass-tagged probe. Two distinct aptamers can recognize target protein monomers and initiate the subsequent hybridization cascade involving binding to the connector probe, formation of an initiator strand, opening of a hairpin probe, and ensuing hybridization with a photocleavable mass-tagged probe. Ultimately, the mass tag was released under ultraviolet light and then subjected to mass spectrometric analysis. In this way, the information regarding the interaction between two protein monomers was successfully converted to the quantitative signal of the mass tag. Using the assay, the expression level of EGFR-HER2 dimer and its relationship with individual protein monomers were determined in four breast cancer cell lines. We are among the first to obtain the absolute level of protein heterodimer, and this quantitative information may be vital in understanding the molecular basis of cancer.
Collapse
Affiliation(s)
- Xiaoxu Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Bo Sun
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.,Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang 222002, China
| | - Jianhua Zhu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Moting Qian
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.,State Key Laboratory of Reproductive Medicine, Nanjing 210029, China.,Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Nanjing 211166, China
| |
Collapse
|
33
|
Maddox AL, Brehove MS, Eliato KR, Saftics A, Romano E, Press MF, Mortimer J, Jones V, Schmolze D, Seewaldt VL, Jovanovic-Talisman T. Molecular Assessment of HER2 to Identify Signatures Associated with Therapy Response in HER2-Positive Breast Cancer. Cancers (Basel) 2022; 14:2795. [PMID: 35681773 PMCID: PMC9179327 DOI: 10.3390/cancers14112795] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/29/2022] [Accepted: 06/01/2022] [Indexed: 11/16/2022] Open
Abstract
Trastuzumab, the prototype HER2-directed therapy, has markedly improved survival for women with HER2-positive breast cancers. However, only 40-60% of women with HER2-positive breast cancers achieve a complete pathological response to chemotherapy combined with HER2-directed therapy. The current diagnostic assays have poor positive-predictive accuracy in identifying therapy-responsive breast cancers. Here, we deployed quantitative single molecule localization microscopy to assess the molecular features of HER2 in a therapy-responsive setting. Using fluorescently labeled trastuzumab as a probe, we first compared the molecular features of HER2 in trastuzumab-sensitive (BT-474 and SK-BR-3) and trastuzumab-resistant (BT-474R and JIMT-1) cultured cell lines. Trastuzumab-sensitive cells had significantly higher detected HER2 densities and clustering. We then evaluated HER2 in pre-treatment core biopsies from women with breast cancer undergoing neoadjuvant therapy. A complete pathological response was associated with a high detected HER2 density and significant HER2 clustering. These results established the nano-organization of HER2 as a potential signature of therapy-responsive disease.
Collapse
Affiliation(s)
- Adam L. Maddox
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (A.L.M.); (M.S.B.); (K.R.E.); (A.S.); (E.R.)
| | - Matthew S. Brehove
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (A.L.M.); (M.S.B.); (K.R.E.); (A.S.); (E.R.)
| | - Kiarash R. Eliato
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (A.L.M.); (M.S.B.); (K.R.E.); (A.S.); (E.R.)
| | - Andras Saftics
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (A.L.M.); (M.S.B.); (K.R.E.); (A.S.); (E.R.)
| | - Eugenia Romano
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (A.L.M.); (M.S.B.); (K.R.E.); (A.S.); (E.R.)
| | - Michael F. Press
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA;
| | - Joanne Mortimer
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Veronica Jones
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Daniel Schmolze
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Victoria L. Seewaldt
- Department of Population Sciences, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Tijana Jovanovic-Talisman
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (A.L.M.); (M.S.B.); (K.R.E.); (A.S.); (E.R.)
| |
Collapse
|
34
|
Bick G, Zhang J, Lower EE, Zhang X. Transcriptional coactivator MED1 in the interface of anti-estrogen and anti-HER2 therapeutic resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:498-510. [PMID: 35800368 PMCID: PMC9255246 DOI: 10.20517/cdr.2022.33] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/30/2022] [Accepted: 04/02/2022] [Indexed: 11/18/2022]
Abstract
Breast cancer is one of the most common cancer and leading causes of death in women in the United States and Worldwide. About 90% of breast cancers belong to ER+ or HER2+ subtypes and are driven by key breast cancer genes Estrogen Receptor and HER2, respectively. Despite the advances in anti-estrogen (endocrine) and anti-HER2 therapies for the treatment of these breast cancer subtypes, unwanted side effects, frequent recurrence and resistance to these treatments remain major clinical challenges. Recent studies have identified ER coactivator MED1 as a key mediator of ER functions and anti-estrogen treatment resistance. Interestingly, MED1 is also coamplified with HER2 and activated by the HER2 signaling cascade, and plays critical roles in HER2-mediated tumorigenesis and response to anti-HER2 treatment as well. Thus, MED1 represents a novel crosstalk point of the HER2 and ER pathways and a highly promising new therapeutic target for ER+ and HER2+ breast cancer treatment. In this review, we will discuss the recent progress on the role of this key ER/HER2 downstream effector MED1 in breast cancer therapy resistance and our development of an innovative RNA nanotechnology-based approach to target MED1 for potential future breast cancer therapy to overcome treatment resistance.
Collapse
Affiliation(s)
- Gregory Bick
- Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jasmine Zhang
- Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Elyse E. Lower
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA. ,University of Cincinnati Cancer Center, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Xiaoting Zhang
- Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.,Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA. ,University of Cincinnati Cancer Center, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.,Correspondence to: Prof. Xiaoting Zhang, Professor and Thomas Boat Endowed Chair, Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267, USA. E-mail:
| |
Collapse
|
35
|
Roviello G, Catalano M, Iannone LF, Marano L, Brugia M, Rossi G, Aprile G, Antonuzzo L. Current status and future perspectives in HER2 positive advanced gastric cancer. Clin Transl Oncol 2022; 24:981-996. [PMID: 35091998 DOI: 10.1007/s12094-021-02760-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 12/13/2021] [Indexed: 02/06/2023]
Abstract
Gastric cancer is one of the most common malignancy worldwide with a prognosis less than 1 year in unresectable or metastatic disease. HER2 expression is the main biomarker to lead the addition of trastuzumab to first line systemic chemotherapy improving the overall survival in advanced HER2-positivegastric adenocarcinoma. The inevitable development of resistance to trastuzumab remains a great problem inasmuch several treatment strategies that have proven effective in breast cancer failed to show clinical benefit in advanced gastric cancer. In this review, we summarize the available data on the mechanisms underlying primary and secondary resistance toHER2-targeted therapy and current challenges in the treatment of HER2-positive advanced gastric cancer refractory to trastuzumab. Further, we describe the prognostic value of new non-invasive screening techniques, the current development of novel agents such us HER2 antibody-drug conjugates and bispecific antibodies, and the strategies with antitumor activity on going.
Collapse
Affiliation(s)
- G Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy.
| | - M Catalano
- School of Human Health Sciences, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - L F Iannone
- Department of Health Science, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - L Marano
- Department of Medical, Surgical and NeuroSciences, Section of Surgery, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena, Italy
| | - M Brugia
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - G Rossi
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - G Aprile
- Department of Oncology, San Bortolo General Hospital, AULSS8 Berica, Vicenza, Italy
| | - L Antonuzzo
- Department of Experimental and Clinical Medicine, University of Florence, 50134, Florence, Italy
- Medical Oncology Unit, Careggi University Hospital, 50134, Florence, Italy
| |
Collapse
|
36
|
Liang H, Zhou L, Chen P, Zheng J, Huang Y, Liang J, Zhong J, Huang Y, Yu M, Guan BO. Optical Microfiber with a Gold Nanorods-Black Phosphorous Nanointerface: An Ultrasensitive Biosensor and Nanotherapy Platform. Anal Chem 2022; 94:8058-8065. [PMID: 35611971 DOI: 10.1021/acs.analchem.2c01499] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The detection and therapy of cancers in the early stage significantly alleviate the associated dangers. Optical devices offer new opportunities for these early measures. However, the clinical translation of the existing methods is severely hindered by their relatively low sensitivity or unclear physiological metabolism. Here, an optical microfiber sensor with a drug loading gold nanorod-black phosphorous nanointerface, as an ultrasensitive biosensor and nanotherapy platform, is developed to meet the early-stage requirement. With interface sensitization and functionalization of the hybrid nanointerface, the microfiber sensor presents an ultrahigh sensing performance, achieving the selective detection of the HER2 biomarker with limits of detection of 0.66 aM in buffer solution and 0.77 aM in 10% serum. It can also distinguish breast cancer cells from other cells in the early stage. Additionally, enabled by the interface, the optical microfiber is able to realize cellular nanotherapy, including photothermal/chemotherapy with pump laser coupling after diagnosis, and evaluate therapy results in real time. The immobilization of the interface on the optical microfiber surface prevents the damage to normal cells induced by nanomaterial enrichment, making the device more efficient and intelligent. This study opens up a new avenue for the development of smart optical platforms for sensitive biosensing and precision therapy.
Collapse
Affiliation(s)
- He Liang
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou 511143, China
| | - Luyan Zhou
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou 511143, China
| | - Pengwei Chen
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou 511143, China
| | - Jiaying Zheng
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou 511143, China
| | - Yunyun Huang
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou 511143, China
| | - Jiaxuan Liang
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou 511143, China
| | - Junyang Zhong
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yugang Huang
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Mingguang Yu
- School of Materials Science and Energy Engineering, Foshan University, Foshan 528000, China
| | - Bai-Ou Guan
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou 511143, China
| |
Collapse
|
37
|
Zou Y, Zheng S, Xie X, Ye F, Hu X, Tian Z, Yan SM, Yang L, Kong Y, Tang Y, Tian W, Xie J, Deng X, Zeng Y, Chen ZS, Tang H, Xie X. N6-methyladenosine regulated FGFR4 attenuates ferroptotic cell death in recalcitrant HER2-positive breast cancer. Nat Commun 2022; 13:2672. [PMID: 35562334 PMCID: PMC9106694 DOI: 10.1038/s41467-022-30217-7] [Citation(s) in RCA: 141] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 04/21/2022] [Indexed: 12/26/2022] Open
Abstract
Intrinsic and acquired anti-HER2 resistance remains a major hurdle for treating HER2-positive breast cancer. Using genome-wide CRISPR/Cas9 screening in vitro and in vivo, we identify FGFR4 as an essential gene following anti-HER2 treatment. FGFR4 inhibition enhances susceptibility to anti-HER2 therapy in resistant breast cancer. Mechanistically, m6A-hypomethylation regulated FGFR4 phosphorylates GSK-3β and activates β-catenin/TCF4 signaling to drive anti-HER2 resistance. Notably, suppression of FGFR4 dramatically diminishes glutathione synthesis and Fe2+ efflux efficiency via the β-catenin/TCF4-SLC7A11/FPN1 axis, resulting in excessive ROS production and labile iron pool accumulation. Ferroptosis, a unique iron-dependent form of oxidative cell death, is triggered after FGFR4 inhibition. Experiments involving patient-derived xenografts and organoids reveals a synergistic effect of anti-FGFR4 with anti-HER2 therapy in breast cancer with either intrinsic or acquired resistance. Together, these results pinpoint a mechanism of anti-HER2 resistance and provide a strategy for overcoming resistance via FGFR4 inhibition in recalcitrant HER2-positive breast cancer.
Collapse
Affiliation(s)
- Yutian Zou
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shaoquan Zheng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xinhua Xie
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Feng Ye
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiaoqian Hu
- School of Biomedical Sciences, Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhi Tian
- College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Shu-Mei Yan
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lu Yang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yanan Kong
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuhui Tang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wenwen Tian
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jindong Xie
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xinpei Deng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yan Zeng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| | - Hailin Tang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Xiaoming Xie
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
38
|
Yang Y, Kozlovskaya V, Zhang Z, Xing C, Zaharias S, Dolmat M, Qian S, Zhang J, Warram JM, Yang ES, Kharlampieva E. Poly( N-vinylpyrrolidone)- block-Poly(dimethylsiloxane)- block-Poly( N-vinylpyrrolidone) Triblock Copolymer Polymersomes for Delivery of PARP1 siRNA to Breast Cancers. ACS APPLIED BIO MATERIALS 2022; 5:1670-1682. [PMID: 35294185 DOI: 10.1021/acsabm.2c00063] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nearly 20% of HER2-positive breast cancers develop resistance to HER2-targeted therapies requiring the use of advanced therapies. Silencing RNA therapy may be a powerful modality for treating resistant HER2 cancers due to its high specificity and low toxicity. However, the systemic administration of siRNAs requires a safe and efficient delivery platform because of siRNA's low stability in physiological fluids, inefficient cellular uptake, immunoreactivity, and rapid clearance. We have developed theranostic polymeric vesicles to overcome these hurdles for encapsulation and delivery of small functional molecules and PARP1 siRNA for in vivo delivery to breast cancer tumors. The 100 nm polymer vesicles were assembled from biodegradable and non-ionic poly(N-vinylpyrrolidone)14-block-poly(dimethylsiloxane)47-block-poly(N-vinylpyrrolidone)14 triblock copolymer PVPON14-PDMS47-PVPON14 using nanoprecipitation and thin-film hydration. We demonstrated that the vesicles assembled from the copolymer covalently tagged with the Cy5.5 fluorescent dye for in vivo imaging could also encapsulate the model drug with high loading efficiency (40%). The dye-loaded vesicles were accumulated in tumors after 18 h circulation in 4TR breast tumor-bearing mice via passive targeting. We found that PARP1 siRNA encapsulated into the vesicles was released intact (13%) into solution by the therapeutic ultrasound treatment as quantified by gel electrophoresis. The PARP1 siRNA-loaded polymersomes inhibited the proliferation of MDA-MB-361TR cells by 34% after 6 days of treatment by suppressing the NF-kB signaling pathway, unlike their scrambled siRNA-loaded counterparts. Finally, the treatment by PARP1 siRNA-loaded vesicles prolonged the survival of the mice bearing 4T1 breast cancer xenografts, with the 4-fold survival increase, unlike the untreated mice after 3 weeks following the treatment. These biodegradable, non-ionic PVPON14-PDMS47-PVPON14 polymeric nanovesicles capable of the efficient encapsulation and delivery of PARP1 siRNA to successfully knock down PARP1 in vivo can provide an advanced platform for the development of precision-targeted therapeutic carriers, which could help develop highly effective drug delivery nanovehicles for breast cancer gene therapy.
Collapse
Affiliation(s)
- Yiming Yang
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Veronika Kozlovskaya
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Zhuo Zhang
- Department of Radiation Oncology, The University of Alabama at Birmingham, Hazelrig Salter Radiation Oncology Center, Birmingham, Alabama 35294, United States.,The O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Chuan Xing
- Department of Radiation Oncology, The University of Alabama at Birmingham, Hazelrig Salter Radiation Oncology Center, Birmingham, Alabama 35294, United States.,The O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Steve Zaharias
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Maksim Dolmat
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Shuo Qian
- Neutron Scattering Division and Second Target Station, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831, United States
| | - Jun Zhang
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Jason M Warram
- The O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States.,Departments of Otolaryngology, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Eddy S Yang
- Department of Radiation Oncology, The University of Alabama at Birmingham, Hazelrig Salter Radiation Oncology Center, Birmingham, Alabama 35294, United States.,The O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States.,Center for Nanoscale Materials and Biointegration, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Eugenia Kharlampieva
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States.,The O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States.,Center for Nanoscale Materials and Biointegration, The University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| |
Collapse
|
39
|
Ligorio F, Zambelli L, Fucà G, Lobefaro R, Santamaria M, Zattarin E, de Braud F, Vernieri C. Prognostic impact of body mass index (BMI) in HER2+ breast cancer treated with anti-HER2 therapies: from preclinical rationale to clinical implications. Ther Adv Med Oncol 2022; 14:17588359221079123. [PMID: 35281350 PMCID: PMC8908398 DOI: 10.1177/17588359221079123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/21/2022] [Indexed: 12/03/2022] Open
Abstract
Human Epidermal growth factor Receptor 2 (HER2) overexpression or HER2 gene amplification defines a subset of breast cancers (BCs) characterized by higher biological and clinical aggressiveness. The introduction of anti-HER2 drugs has remarkably improved clinical outcomes in patients with both early-stage and advanced HER2+ BC. However, some HER2+ BC patients still have unfavorable outcomes despite optimal anti-HER2 therapies. Retrospective clinical analyses indicate that overweight and obesity can negatively affect the prognosis of patients with early-stage HER2+ BC. This association could be mediated by the interplay between overweight/obesity, alterations in systemic glucose and lipid metabolism, increased systemic inflammatory status, and the stimulation of proliferation pathways resulting in the stimulation of HER2+ BC cell growth and resistance to anti-HER2 therapies. By contrast, in the context of advanced disease, a few high-quality studies, which were included in a meta-analysis, showed an association between high body mass index (BMI) and better clinical outcomes, possibly reflecting the negative prognostic role of malnourishment and cachexia in this setting. Of note, overweight and obesity are modifiable factors. Therefore, uncovering their prognostic role in patients with early-stage or advanced HER2+ BC could have clinical relevance in terms of defining subsets of patients requiring more or less aggressive pharmacological treatments, as well as of designing clinical trials to investigate the therapeutic impact of lifestyle interventions aimed at modifying body weight and composition. In this review, we summarize and discuss the available preclinical evidence supporting the role of adiposity in modulating HER2+ BC aggressiveness and resistance to therapies, as well as clinical studies reporting on the prognostic role of BMI in patients with early-stage or advanced HER2+ BC.
Collapse
Affiliation(s)
- Francesca Ligorio
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Luca Zambelli
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanni Fucà
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Riccardo Lobefaro
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marzia Santamaria
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy
| | - Emma Zattarin
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Claudio Vernieri
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy
| |
Collapse
|
40
|
Wang P, Mak VCY, Cheung LWT. Drugging IGF-1R in cancer: New insights and emerging opportunities. Genes Dis 2022; 10:199-211. [PMID: 37013053 PMCID: PMC10066341 DOI: 10.1016/j.gendis.2022.03.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/02/2022] [Indexed: 11/19/2022] Open
Abstract
The insulin-like growth factor (IGF) axis plays important roles in cancer development and metastasis. The type 1 IGF receptor (IGF-1R) is a key member in the IGF axis and has long been recognized for its oncogenic role in multiple cancer lineages. Here we review the occurrence of IGF-1R aberrations and activation mechanisms in cancers, which justify the development of anti-IGF-1R therapies. We describe the therapeutic agents available for IGF-1R inhibition, with focuses on the recent or ongoing pre-clinical and clinical studies. These include antisense oligonucleotide, tyrosine kinase inhibitors and monoclonal antibodies which may be conjugated with cytotoxic drug. Remarkably, simultaneous targeting of IGF-1R and several other oncogenic vulnerabilities has shown early promise, highlighting the potential benefits of combination therapy. Further, we discuss the challenges in targeting IGF-1R so far and new concepts to improve therapeutic efficacy such as blockage of the nuclear translocation of IGF-1R.
Collapse
|
41
|
Recent advances and limitations in the application of kahalalides for the control of cancer. Biomed Pharmacother 2022; 148:112676. [PMID: 35149387 PMCID: PMC9004612 DOI: 10.1016/j.biopha.2022.112676] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/19/2022] [Accepted: 01/22/2022] [Indexed: 11/24/2022] Open
Abstract
Since the discovery of the kahalalide family of marine depsipeptides in 1993, considerable work has been done to develop these compounds as new and biologically distinct anti-cancer agents. Clinical trials and laboratory research have yielded a wealth of data that indicates tolerance of kahalalides in healthy cells and selective activity against diseased cells. Currently, two molecules have attracted the greates level of attention, kahalalide F (KF) and isokahalalide F (isoKF, Irvalec, PM 02734, elisidepsin). Both compounds were originally isolated from the sarcoglossan mollusk Elysia rufescens but due to distinct structural characteristics it has been hypothesized and recently shown that the ultimate origin of the molecules is microbial. The search for their true source has been a subject of considerable research in the anticipation of finding new analogs and a culturable expression system that can produce sufficient material through fermentation to be industrially relevant.
Collapse
|
42
|
East MP, Johnson GL. Adaptive chromatin remodeling and transcriptional changes of the functional kinome in tumor cells in response to targeted kinase inhibition. J Biol Chem 2021; 298:101525. [PMID: 34958800 PMCID: PMC8888345 DOI: 10.1016/j.jbc.2021.101525] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/06/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022] Open
Abstract
Pharmacological inhibition of protein kinases induces adaptive reprogramming of tumor cell regulatory networks by altering expression of genes that regulate signaling, including protein kinases. Adaptive responses are dependent on transcriptional changes resulting from remodeling of enhancer and promoter landscapes. Enhancer and promoter remodeling in response to targeted kinase inhibition is controlled by changes in open chromatin state and by activity of specific transcription factors, such as c-MYC. This review focuses on the dynamic plasticity of protein kinase expression of the tumor cell kinome and the resulting adaptive resistance to targeted kinase inhibition. Plasticity of the functional kinome has been shown in patient window trials where triple-negative and human epidermal growth factor receptor 2–positive breast cancer patient tumors were characterized by RNAseq after biopsies before and after 1 week of therapy. The expressed kinome changed dramatically during drug treatment, and these changes in kinase expression were shown in cell lines and xenografts in mice to be correlated with adaptive tumor cell drug resistance. The dynamic transcriptional nature of the kinome also differs for inhibitors targeting different kinase signaling pathways (e.g., BRAF-MEK-ERK versus PI3K-AKT) that are commonly activated in cancers. Heterogeneity arising from differences in gene regulation and mutations represents a challenge to therapeutic durability and prevention of clinical drug resistance with drug-tolerant tumor cell populations developing and persisting through treatment. We conclude that understanding the heterogeneity of kinase expression at baseline and in response to therapy is imperative for development of combinations and timing intervals of therapies making interventions durable.
Collapse
Affiliation(s)
- Michael P East
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Gary L Johnson
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.
| |
Collapse
|
43
|
Fatima I, Rahdar A, Sargazi S, Barani M, Hassanisaadi M, Thakur VK. Quantum Dots: Synthesis, Antibody Conjugation, and HER2-Receptor Targeting for Breast Cancer Therapy. J Funct Biomater 2021; 12:75. [PMID: 34940554 PMCID: PMC8708439 DOI: 10.3390/jfb12040075] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is becoming one of the main lethal carcinomas in the recent era, and its occurrence rate is increasing day by day. There are different breast cancer biomarkers, and their overexpression takes place in the metastasis of cancer cells. The most prevalent breast cancer biomarker is the human epidermal growth factor receptor2 (HER2). As this biomarker is overexpressed in malignant breast tissues, it has become the main focus in targeted therapies to fight breast cancer. There is a cascade of mechanisms involved in metastasis and cell proliferation in cancer cells. Nanotechnology has become extremely advanced in targeting and imaging cancerous cells. Quantum dots (QDs) are semiconductor NPs, and they are used for bioimaging, biolabeling, and biosensing. They are synthesized by different approaches such as top-down, bottom-up, and synthetic methods. Fully human monoclonal antibodies synthesized using transgenic mice having human immunoglobulin are used to target malignant cells. For the HER2 receptor, herceptin® (trastuzumab) is the most specific antibody (Ab), and it is conjugated with QDs by using different types of coupling mechanisms. This quantum dot monoclonal antibody (QD-mAb) conjugate is localized by injecting it into the blood vessel. After the injection, it goes through a series of steps to reach the intracellular space, and bioimaging of specifically the HER2 receptor occurs, where apoptosis of the cancer cells takes place either by the liberation of Ab or the free radicals.
Collapse
Affiliation(s)
- Iqra Fatima
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 98167-43463, Iran;
| | - Mahmood Barani
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman 76169-13555, Iran; (M.B.); (M.H.)
| | - Mohadeseh Hassanisaadi
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman 76169-13555, Iran; (M.B.); (M.H.)
- Department of Plant Protection, Shahid Bahonar University of Kerman, Kerman 76184-11764, Iran
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Center, SRUC, Edinburgh EH9 3JG, UK
- School of Engineering, University of Petroleum & Energy Studies (UPES), Dehradun 248007, Uttarakhand, India
| |
Collapse
|
44
|
Basile D, Simionato F, Cappetta A, Garattini SK, Roviello G, Aprile G. State-of-the-Art of Monoclonal Antibodies for the Treatment of Gastric Cancer. Biologics 2021; 15:451-462. [PMID: 34764633 PMCID: PMC8572727 DOI: 10.2147/btt.s290323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/05/2021] [Indexed: 12/07/2022]
Abstract
Gastric cancer (GC) is a complex and heterogeneous disease with poor prognosis and limited available treatment options. During recent years, several molecular stratifications have been proposed to optimize the overall treatment strategy for GC patients. Breakthroughs in cancer biology and in molecular profiling through DNA and RNA sequencing are now opening novel landscapes, leading to the personalization of molecular matched therapy. In particular, therapies against HER2, Claudine 18.2, Fibroblast Growth Factor Receptors (FGFR), and other molecular alterations could significantly improve survival outcomes in the advance phase of the disease. Furthermore, immunotherapy with checkpoint inhibitors also represents a promising option in a selected population. Hoping that precision oncology will enter soon in clinical practice, our review describes the state of the art of many novel pathways and the current evidence supporting the use of monoclonal antibodies implicated in GC treatment.
Collapse
Affiliation(s)
- Debora Basile
- Department of Oncology, San Bortolo General Hospital, AULSS8 Berica, Vicenza, Italy
| | - Francesca Simionato
- Department of Oncology, San Bortolo General Hospital, AULSS8 Berica, Vicenza, Italy
| | - Alessandro Cappetta
- Department of Oncology, San Bortolo General Hospital, AULSS8 Berica, Vicenza, Italy
| | | | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, AULSS8 Berica, Vicenza, Italy
| |
Collapse
|
45
|
Salkeni MA, Rizvi W, Hein K, Higa GM. Neu Perspectives, Therapies, and Challenges for Metastatic HER2-Positive Breast Cancer. BREAST CANCER-TARGETS AND THERAPY 2021; 13:539-557. [PMID: 34602823 PMCID: PMC8481821 DOI: 10.2147/bctt.s288344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/20/2021] [Indexed: 12/26/2022]
Abstract
Even though gene amplification or protein overexpression occurs in approximately one-fifth of all breast cancers, the discovery of HER2 has, nevertheless, had profound implications for the disease. Indeed, the characterization of the receptor resulted in a number of significant advances. Structurally, unique features provided avenues for the development of numerous compounds with target-specificity; molecularly, biological constructs revealed a highly complex, internal signal transduction pathway with regulatory effects on tumor proliferation, survival, and perhaps, even resistance; and clinically, disease outcomes manifested its predictive and prognostic value. Yet despite the receptor’s utility, the beneficial effects are diminished by tumor recurrence after neo- or adjuvant therapy as well as losses resulting from the inability to cure patients with metastatic disease. What these observations suggest is that while tumor response may be partially linked to uncoupling cell surface message reception and nuclear gene expression, as well as recruitment of the innate immune system, disease progression and/or resistance may involve a reprogrammable signaling mainframe that elicits alternative growth and survival signals. This review attempts to meld current perceptions related to HER2-positive metastatic breast cancer with particular attention to current biological insights and therapeutic challenges.
Collapse
Affiliation(s)
- Mohamad Adham Salkeni
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wajeeha Rizvi
- Department of Internal Medicine, West Virginia University, Morgantown, WV, USA
| | - Kyaw Hein
- Department of Business, Lamar University, Houston, TX, USA
| | - Gerald M Higa
- Departments of Clinical Pharmacy and Medicine, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
46
|
Pareri AU, Koijam AS, Kumar C. Breaking the Silence of Tumor Response: Future Prospects of Targeted Radionuclide Therapy. Anticancer Agents Med Chem 2021; 22:1845-1858. [PMID: 34477531 DOI: 10.2174/1871520621666210903152354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/12/2021] [Accepted: 07/19/2021] [Indexed: 01/10/2023]
Abstract
Therapy-induced tumor resistance has always been a paramount hurdle in the clinical triumph of cancer therapy. Resistance acquired by tumor through interventions of chemotherapeutic drugs, ionizing radiation, and immunotherapy in the patientsis a severe drawback and major cause of recurrence of tumor and failure of therapeutic responses. To counter acquired resistance in tumor cells, several strategies are practiced such as chemotherapy regimens, immunotherapy, and immunoconjugates, but the outcome is very disappointing for the patients as well as clinicians. Radionuclide therapy using alpha or beta-emitting radionuclide as payload became state-of-the-art for cancer therapy. With the improvement in dosimetric studies, development of high-affinity target molecules, and design of several novel chelating agents which provide thermodynamically stable complexes in vivo, the scope of radionuclide therapy has increased by leaps and bounds. Additionally, radionuclide therapy along with the combination of chemotherapy is gaining importance in pre-clinics, which is quite encouraging. Thus, it opens an avenue for newer cancer therapy modalities where chemotherapy, radiation therapy, and immunotherapy are unable to break the silence of tumor response. This article describes, in brief, the causes of tumor resistance and discusses the potential of radionuclide therapy to enhance tumor response.
Collapse
Affiliation(s)
| | | | - Chandan Kumar
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre Mumbai-400085, India
| |
Collapse
|
47
|
Randomized Phase II Trial of Capecitabine and Lapatinib with or without IMC-A12 (Cituxumumab) in Patients with HER2-Positive Advanced Breast Cancer Previously Treated with Trastuzumab and Chemotherapy: NCCTG N0733 (Alliance). Breast Cancer Res Treat 2021; 188:477-487. [PMID: 33852121 PMCID: PMC8262517 DOI: 10.1007/s10549-021-06221-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/30/2021] [Indexed: 02/01/2023]
Abstract
PURPOSE To compare efficacy and safety of capecitabine and lapatinib with or without IMC-A12 (cituxumumab) in patients with HER2-positive metastatic breast cancer (MBC) previously treated with trastuzumab. PATIENTS AND METHODS Following an initial safety run-in cohort, patients were randomized 1:2 to Arm A (capecitabine and lapatinib) or to Arm B (capecitabine, lapatinib, and cituxumumab). Given the frequency of non-hematologic grade ≥ 3 adverse events in those receiving the three-drug combination in the safety cohort, lapatinib and capecitabine doses were reduced in Arm B only. The primary objective was to determine if the addition of cituxumumab to capecitabine and lapatinib improved progression-free survival (PFS) compared with capecitabine and lapatinib. Secondary objectives included a comparison between arms of other clinical endpoints, safety, change in overall quality of life (QOL) and self-assessed fatigue, rash, diarrhea, and hand-foot syndrome. RESULTS From July 2008 to March 2012, 68 patients (out of 142 planned) were enrolled and 63 were evaluable, including 8 for the safety run-in and 55 for the randomized cohort. Study enrollment was stopped early due to slow accrual. The addition of cituxumumab to capecitabine and lapatinib did not improve PFS (HR 0.93, 95% CI: 0.52-1.64). Furthermore, no difference in objective response rate or overall survival (OS) was observed. No difference between arms was observed in grade ≥ 3 adverse events, overall QOL change from baseline after 4 cycles of treatment. CONCLUSION The addition of cituxumumab to lapatinib and capecitabine did not improve PFS or OS compared with lapatinib and capecitabine in patients with HER2-positive MBC. CLINICAL TRIAL REGISTRY ClinicalTrials.gov Identifier: NCT00684983.
Collapse
|
48
|
Roviello G, Aprile G, D'Angelo A, Iannone LF, Roviello F, Polom K, Mini E, Catalano M. Human epidermal growth factor receptor 2 (HER2) in advanced gastric cancer: where do we stand? Gastric Cancer 2021; 24:765-779. [PMID: 33742317 DOI: 10.1007/s10120-021-01182-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
Gastric cancer is one of the most common malignancy worldwide. In unresectable or metastatic disease, the prognosis is poor and in generally less than a year. HER2 expression remains an important biomarker to lead the addition of trastuzumab to first-line systemic chemotherapy in unresectable or metastatic gastroesophageal adenocarcinoma. To date, a major issue is represented by resistance to trastuzumab developed during treatment, considering the not improved outcomes in this molecular subtype of gastroesophageal adenocarcinoma to other HER2 target strategies. In this review, we summarize the available data on the mechanisms underlying primary and secondary resistance to HER2-targeted therapy and current challenges in the treatment of HER2-positive advanced gastric cancer refractory to trastuzumab. Furthermore, we describe the prognostic value of new non-invasive screening methods, under development novel agents (e.g., HER2 antibody-drug conjugates and bispecific antibodies) and strategies with antitumor activity in early studies.
Collapse
Affiliation(s)
- Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy.
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, Vicenza, Italy
| | - Alberto D'Angelo
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK
| | | | - Franco Roviello
- Department of Medical, Surgical and Neuro Sciences, Section of Surgery, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena, Italy
| | - Karol Polom
- Department of Medical, Surgical and Neuro Sciences, Section of Surgery, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena, Italy
- Department of Surgical Oncology, Gdansk Medical University, Gdansk, Poland
| | - Enrico Mini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy
| | - Martina Catalano
- School of Human Health Sciences, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| |
Collapse
|
49
|
Zhu M, Wang DD, Yan H. Genotype-determined EGFR-RTK heterodimerization and its effects on drug resistance in lung Cancer treatment revealed by molecular dynamics simulations. BMC Mol Cell Biol 2021; 22:34. [PMID: 34112110 PMCID: PMC8191231 DOI: 10.1186/s12860-021-00358-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/10/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) and its signaling pathways play a vital role in pathogenesis of lung cancer. By disturbing EGFR signaling, mutations of EGFR may lead to progression of cancer or the emergence of resistance to EGFR-targeted drugs. RESULTS We investigated the correlation between EGFR mutations and EGFR-receptor tyrosine kinase (RTK) crosstalk in the signaling network, in order to uncover the drug resistance mechanism induced by EGFR mutations. For several EGFR wild type (WT) or mutated proteins, we measured the EGFR-RTK interactions using several computational methods based on molecular dynamics (MD) simulations, including geometrical characterization of the interfaces and conventional estimation of free energy of binding. Geometrical properties, namely the matching rate of atomic solid angles in the interfaces and center-of-mass distances between interacting atoms, were extracted relying on Alpha Shape modeling. For a couple of RTK partners (c-Met, ErbB2 and IGF-1R), results have shown a looser EGFR-RTK crosstalk for the drug-sensitive EGFR mutant while a tighter crosstalk for the drug-resistant mutant. It guarantees the genotype-determined EGFR-RTK crosstalk, and further proposes a potential drug resistance mechanism by amplified EGFR-RTK crosstalk induced by EGFR mutations. CONCLUSIONS This study will lead to a deeper understanding of EGFR mutation-induced drug resistance mechanisms and promote the design of innovative drugs.
Collapse
Affiliation(s)
- Mengxu Zhu
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong.
| | - Debby D Wang
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Hong Yan
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
50
|
Angus SP, Stuhlmiller TJ, Mehta G, Bevill SM, Goulet DR, Olivares-Quintero JF, East MP, Tanioka M, Zawistowski JS, Singh D, Sciaky N, Chen X, He X, Rashid NU, Chollet-Hinton L, Fan C, Soloway MG, Spears PA, Jefferys S, Parker JS, Gallagher KK, Forero-Torres A, Krop IE, Thompson AM, Murthy R, Gatza ML, Perou CM, Earp HS, Carey LA, Johnson GL. FOXA1 and adaptive response determinants to HER2 targeted therapy in TBCRC 036. NPJ Breast Cancer 2021; 7:51. [PMID: 33980863 PMCID: PMC8115531 DOI: 10.1038/s41523-021-00258-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 04/08/2021] [Indexed: 12/11/2022] Open
Abstract
Inhibition of the HER2/ERBB2 receptor is a keystone to treating HER2-positive malignancies, particularly breast cancer, but a significant fraction of HER2-positive (HER2+) breast cancers recur or fail to respond. Anti-HER2 monoclonal antibodies, like trastuzumab or pertuzumab, and ATP active site inhibitors like lapatinib, commonly lack durability because of adaptive changes in the tumor leading to resistance. HER2+ cell line responses to inhibition with lapatinib were analyzed by RNAseq and ChIPseq to characterize transcriptional and epigenetic changes. Motif analysis of lapatinib-responsive genomic regions implicated the pioneer transcription factor FOXA1 as a mediator of adaptive responses. Lapatinib in combination with FOXA1 depletion led to dysregulation of enhancers, impaired adaptive upregulation of HER3, and decreased proliferation. HER2-directed therapy using clinically relevant drugs (trastuzumab with or without lapatinib or pertuzumab) in a 7-day clinical trial designed to examine early pharmacodynamic response to antibody-based anti-HER2 therapy showed reduced FOXA1 expression was coincident with decreased HER2 and HER3 levels, decreased proliferation gene signatures, and increased immune gene signatures. This highlights the importance of the immune response to anti-HER2 antibodies and suggests that inhibiting FOXA1-mediated adaptive responses in combination with HER2 targeting is a potential therapeutic strategy.
Collapse
Affiliation(s)
- Steven P Angus
- Department of Pharmacology, UNC Chapel Hill, Chapel Hill, NC, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Gaurav Mehta
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Samantha M Bevill
- Department of Pharmacology, UNC Chapel Hill, Chapel Hill, NC, USA
- Massachusetts General Hospital, Cambridge, MA, USA
| | - Daniel R Goulet
- Department of Pharmacology, UNC Chapel Hill, Chapel Hill, NC, USA
- Koch Institute, Massachusetts Institute of Technology, Boston, MA, USA
| | | | - Michael P East
- Department of Pharmacology, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Maki Tanioka
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
- Hyogo Cancer Center, Akashi, Japan
| | | | - Darshan Singh
- Department of Pharmacology, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Noah Sciaky
- Department of Pharmacology, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Xin Chen
- Department of Pharmacology, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Xiaping He
- Department of Genetics, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Naim U Rashid
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
- Department of Biostatistics, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Lynn Chollet-Hinton
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Cheng Fan
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Matthew G Soloway
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Patricia A Spears
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Stuart Jefferys
- Department of Genetics, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Joel S Parker
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Kristalyn K Gallagher
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
- Department of Surgery, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Andres Forero-Torres
- University of Alabama-Birmingham School of Medicine, Birmingham, AL, USA
- Seattle Genetics, Inc., Seattle, WA, USA
| | - Ian E Krop
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alastair M Thompson
- Department of Breast Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
- Baylor College of Medicine, Houston, TX, USA
| | - Rashmi Murthy
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Michael L Gatza
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Charles M Perou
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, UNC Chapel Hill, Chapel Hill, NC, USA
| | - H Shelton Earp
- Department of Pharmacology, UNC Chapel Hill, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
- Department of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Lisa A Carey
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
- Department of Medicine, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Gary L Johnson
- Department of Pharmacology, UNC Chapel Hill, Chapel Hill, NC, USA.
- UNC Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|