1
|
Babaei Khorzoughi S, Tavakoli M, Mortazavi M, Jafarnejad Z, Malekpour A, Kopaiee Malek T, Kargar F. A review of recombinant HER3 affibodies with an effective diagnostic view of cancer cells. J Drug Target 2025; 33:316-327. [PMID: 39485069 DOI: 10.1080/1061186x.2024.2420202] [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: 07/05/2024] [Revised: 09/10/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024]
Abstract
Breast cancer is one of the leading causes of cancer-related deaths among women globally. Factors like increased expression of HER family members contribute to its development, with elevated HER3 levels-especially in conjunction with tyrosine kinase receptors like HER2-playing a critical role in activating cancer pathways essential for cell survival and proliferation. Detecting high HER3 levels is vital for effective treatment. Affibody proteins, a class that includes antibodies, are used to identify elevated HER3 expression due to their high binding affinity. These innovative non-immune probes show promise in therapy, diagnostics, and biotechnology because of their exceptional specificity and affinity for target proteins. The design of recombinant affibodies enhances HER3 detection accuracy and supports the development of targeted therapies. Advanced engineering techniques optimize these affibodies for stability and binding efficacy, making them suitable for clinical applications. Additionally, their versatility allows integration with imaging technologies for real-time monitoring of HER3 expression and therapeutic responses. This comprehensive approach could lead to more personalized treatment options for patients with HER3-positive breast cancers, improving patient management and outcomes. This study presents recombinant affibodies designed to bind HER3 for cancer cell identification and introduces novel methods for producing various affibody molecules.
Collapse
Affiliation(s)
- Sahar Babaei Khorzoughi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Mehrnoosh Tavakoli
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Mojtaba Mortazavi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Zahra Jafarnejad
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | | | - Tara Kopaiee Malek
- Department of Cell and Molecular Biology, Faculty of Science, Azad University of Damghan, Damghan, Iran
| | - Farzane Kargar
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Geng X, Azarbarzin S, Yang Z, Lapidus RG, Fan X, Teng Y, Mehra R, Cullen KJ, Dan H. Evaluation of co‑inhibition of ErbB family kinases and PI3K for HPV‑negative head and neck squamous cell carcinoma. Oncol Rep 2025; 53:38. [PMID: 39886949 PMCID: PMC11800064 DOI: 10.3892/or.2025.8871] [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: 03/05/2024] [Accepted: 10/29/2024] [Indexed: 02/01/2025] Open
Abstract
The ErbB/HER family of protein‑tyrosine kinases and PI3K represent crucial targets in the treatment of head and neck squamous cell carcinoma (HNSCC). A combination therapy of afatinib (ErbB inhibitor) and copanlisib (PI3K inhibitor), both Food and Drug Administration‑approved kinase inhibitors, can suppress the growth of human papillomavirus (HPV)‑positive HNSCC. The current study further evaluated the efficacy and clinical potential of this combination therapy for the treatment of HPV‑negative HNSCC in vitro and in vivo. Sulforhodamine B cell viability assay and Annexin V/propidium iodide staining demonstrated that this combination treatment markedly enhanced inhibition of cell viability and reduced cell survival when compared with treatment with either inhibitor alone in two HPV‑negative HNSCC cell lines. Notably, this combination also led to significant inhibition of xenograft tumor growth in mice, without any apparent effects on body weight. Western blot analysis found that copanlisib alone effectively blocked PI3K/Akt signaling but caused upregulation of HER2 and HER3 phosphorylation, as reported in other types of cancer. However, the combination of copanlisib and afatinib completely blocked phosphorylation of the ErbB family (including HER3) and Akt, while also increasing apoptosis. In conclusion, these results suggested that co‑targeting the ErbB family kinases and PI3K using a combination treatment of afatinib and copanlisib may have clinical potential for patients with HPV‑negative HNSCC.
Collapse
Affiliation(s)
- Xinyan Geng
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shirin Azarbarzin
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zejia Yang
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rena G. Lapidus
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xiaoxuan Fan
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Ranee Mehra
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kevin J. Cullen
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hancai Dan
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
3
|
Trinder A, Ding K, Zhang J. The Therapeutic Significance of HER3 in Non-small Cell Lung Cancer (NSCLC): A Review Study. Curr Med Chem 2025; 32:434-446. [PMID: 38231075 DOI: 10.2174/0109298673269305231115102542] [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: 06/27/2023] [Revised: 09/07/2023] [Accepted: 10/08/2023] [Indexed: 01/18/2024]
Abstract
HER3 (Human Epidermal Growth Factor Receptor 3) is frequently overexpressed in various cancers, including non-small cell lung cancer (NSCLC), with a prevalence of 83% in primary tumors. Its involvement in tumorigenesis and resistance to targeted therapies makes HER3 a promising target for cancer treatment. Despite being initially considered "undruggable" due to its lack of catalytic activity, significant progress has been made in the development of anti-HER3 therapeutics. Monoclonal antibodies such as lumretuzumab, seribantumab, and patritumab have shown potential in targeting HER3 to overcome resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). Additionally, antibody-drug conjugates (ADCs) like HER3-DXd (patritumab deruxtecan) are new drug candidates that have demonstrated selective delivery of cytotoxic chemicals to NSCLC cells by exploiting HER3's widespread expression, minimizing cytotoxicity. This review aims to evaluate the efficacy of current HER3 therapeutics in development and their therapeutic potential in NSCLC, incorporating evidence from clinical trials.
Collapse
Affiliation(s)
- Amelia Trinder
- Hatherly Laboratories, Faculty of Health and Life Sciences, Medical School, Institute of Biomedical and Clinical Sciences, University of Exeter, Streatham Campus, Exeter EX4 4PS, UK
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Jinwei Zhang
- Hatherly Laboratories, Faculty of Health and Life Sciences, Medical School, Institute of Biomedical and Clinical Sciences, University of Exeter, Streatham Campus, Exeter EX4 4PS, UK
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| |
Collapse
|
4
|
Mihaylova R, Momekova D, Elincheva V, Momekov G. Immunoconjugates as an Efficient Platform for Drug Delivery: A Resurgence of Natural Products in Targeted Antitumor Therapy. Pharmaceuticals (Basel) 2024; 17:1701. [PMID: 39770542 PMCID: PMC11677665 DOI: 10.3390/ph17121701] [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: 11/14/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
The present review provides a detailed and comprehensive discussion on antibody-drug conjugates (ADCs) as an evolving new modality in the current therapeutic landscape of malignant diseases. The principle concepts of targeted delivery of highly toxic agents forsaken as stand-alone drugs are examined in detail, along with the biochemical and technological tools for their successful implementation. An extensive analysis of ADCs' major components is conducted in parallel with their function and impact on the stability, efficacy, safety, and resistance profiles of the immunoconjugates. The scope of the article covers the major classes of currently validated natural compounds used as payloads, with an emphasis on their structural and mechanistic features, natural origin, and distribution. Future perspectives in ADCs' design are thoroughly explored, addressing their inherent or emerging challenges and limitations. The survey also provides a comprehensive overview of the molecular rationale for active tumor targeting of ADC-based platforms, exploring the cellular biology and clinical relevance of validated tumor markers used as a "homing" mechanism in both hematological and solid tumor malignancies.
Collapse
Affiliation(s)
- Rositsa Mihaylova
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| | - Denitsa Momekova
- Department “Pharmaceutical Technology and Biopharmaceutics”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria;
| | - Viktoria Elincheva
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| | - Georgi Momekov
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| |
Collapse
|
5
|
Martins SA, Correia JDG. 99mTc(I)-Labeled His-Tagged Proteins: Impact in the Development of Novel Imaging Probes and in Drug Discovery. Chembiochem 2024; 25:e202400645. [PMID: 39158861 DOI: 10.1002/cbic.202400645] [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: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 08/20/2024]
Abstract
Technetium-99 m (99mTc) remains the cornerstone of nuclear medicine for single photon emission computed tomography (SPECT) due to its widespread availability and chemical and physical features. Its multiple oxidation states allow for the design and production of radiopharmaceuticals with versatile properties, namely in terms of pharmacokinetic profile. 99mTc(V) is the most common oxidation state, but 99mTc(I) gained traction after the pioneering work of Alberto and colleagues, which resulted in the introduction of the organometallic core fac-[99mTc(CO)3(H2O)3]+. This core is readily available from [99mTcO4]- and displays three labile water molecules that can be easily swapped for ligands with different denticity and/or donor atoms in aqueous environment. This makes it possible to radiolabel small molecules as well as high molecular weight molecules, such as antibodies or other proteins, while assuring biological activity. Direct radiolabelling of those proteins with fac-[99mTc(CO)3]+ under mild conditions is accomplished through incorporation of a polyhistidine tag (His-tag), a commonly used tag for purification of recombinant proteins. This review aims to address the direct radiolabelling of His-tagged macromolecules with fac-[99mTc(CO)3]+ for development of molecular imaging agents and the impact of this technology in the discovery and development of imaging and/or therapeutic agents towards clinical application.
Collapse
Affiliation(s)
- Sofia A Martins
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal
| | - João D G Correia
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal
| |
Collapse
|
6
|
Jiang Z, Gu Z, Yu X, Cheng T, Liu B. Research progress on the role of bypass activation mechanisms in resistance to tyrosine kinase inhibitors in non-small cell lung cancer. Front Oncol 2024; 14:1447678. [PMID: 39582541 PMCID: PMC11581962 DOI: 10.3389/fonc.2024.1447678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/25/2024] [Indexed: 11/26/2024] Open
Abstract
The clinical application of small molecule tyrosine kinase inhibitors (TKIs) has significantly improved the quality of life and prognosis of patients with non-small cell lung cancer (NSCLC) carrying driver genes. However, resistance to TKI treatment is inevitable. Bypass signal activation is one of the important reasons for TKI resistance. Although TKI drugs inhibit downstream signaling pathways of driver genes, key signaling pathways within tumor cells can still be persistently activated through bypass routes such as MET gene amplification, EGFR gene amplification, and AXL activation. This continuous activation maintains tumor cell growth and proliferation, leading to TKI resistance. The fundamental strategy to treat TKI resistance mediated by bypass activation involves simultaneously inhibiting the activated bypass signals and the original driver gene signaling pathways. Some clinical trials based on this combined treatment approach have yielded promising preliminary results, offering more treatment options for NSCLC patients with TKI resistance. Additionally, early identification of resistance mechanisms through liquid biopsy, personalized targeted therapy against these mechanisms, and preemptive targeting of drug-tolerant persistent cells may provide NSCLC patients with more sustained and effective treatment.
Collapse
Affiliation(s)
- Ziyang Jiang
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihan Gu
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaomin Yu
- Department of Emergency Medicine, West China Hospital, Sichuan University, West China School of Nursing, Sichuan University, Chengdu, China
- Institute of Disaster Medicine, Sichuan University, Chengdu, China
- Nursing Key Laboratory of Sichuan Province, West China Hospital, Chengdu, China
| | - Tao Cheng
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Bofu Liu
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Bermúdez-Abreut E, Bergado Báez G, Martínez Pestano M, Attanasio G, Gonzales Castillo CY, Hernández Fernández DR, Alvarez-Arzola R, Alimonti A, Sánchez-Ramírez B. Antitumor activity of PAbs generated by immunization with a novel HER3-targeting protein-based vaccine candidate in preclinical models. Front Oncol 2024; 14:1472607. [PMID: 39479017 PMCID: PMC11521786 DOI: 10.3389/fonc.2024.1472607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Despite the cumulative evidence supporting HER3 as a target for antitumor therapies, no agents targeting HER3 have been approved for cancer treatment. Most of the agents evaluated in preclinical and clinical trials have been specific monoclonal antibodies (MAbs), with few examples of active immunotherapy directed against this receptor. However, some cancer vaccine formats may generate polyclonal antibodies (PAbs) that replicate the diverse effector mechanisms of MAbs, including ligand neutralization and receptor degradation. In this study, we developed a protein subunit-based monovalent vaccine candidate targeting the extracellular domain (ECD) of HER3. Immunization of mice with a formulation targeting murine ErbB3-ECD successfully overcome tolerance to this self-antigen, inducing high titers of ErbB3-specific PAbs. The antitumor potential of this formulation and the induced PAbs was demonstrated in vivo and in vitro in an ErbB3-overexpressing 3LL-D122-derived tumor model. The immunogenicity of the HER3-ECD-based vaccine candidate was confirmed by the induction of high titers of HER3-specific PAbs. Consistent with the initial results, HER3-ECD-targeting PAbs were cytotoxic in several human epithelial tumor cell lines and exerted antitumor effects in vivo. These results support the value of HER3 as a tumor antigen and the effector mechanisms of HER3-specific therapeutic MAbs, while suggesting the potential of the proposed vaccine candidate for the treatment of HER3-expressing carcinomas.
Collapse
Affiliation(s)
| | - Gretchen Bergado Báez
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | | | - Giuseppe Attanasio
- Department of Molecular Oncology, Institute of Oncology Research (IOR), Bellinzona, Switzerland
| | | | | | - Rydell Alvarez-Arzola
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Andrea Alimonti
- Department of Molecular Oncology, Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Faculty of Medicine, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Medicine, University of Padua, Padua, Italy
- Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | | |
Collapse
|
8
|
Soo RA, Clinthorne G, Santhanagopal A, Wu C, Karnoub M, Patel P, Bubendorf L. HER3 is widely expressed across diverse subtypes of NSCLC in a retrospective analysis of archived tissue samples. Future Oncol 2024; 20:2961-2970. [PMID: 39320886 PMCID: PMC11572136 DOI: 10.1080/14796694.2024.2398983] [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: 05/01/2024] [Accepted: 08/28/2024] [Indexed: 09/26/2024] Open
Abstract
Aim: This noninterventional study (NCT05769764) aimed to characterize human epidermal growth factor receptor 3 (HER3) expression in non-small cell lung cancer (NSCLC) by patient, clinical or tumor characteristics.Methods: HER3 immunohistochemistry was performed in archival tissue samples from patients with advanced or metastatic NSCLC. Samples were scored for membrane percent positivity and intensity. Membrane H-scores were calculated.Results: Of 203 evaluable samples, HER3 expression was observed in 98.5%, including all histologies, genomic subtypes and regardless of prior systemic anticancer treatments. The median H-score was 140, and 70.4% had a HER3 intensity of 3+.Conclusion: HER3 is widely expressed in NSCLC, indicating that HER3-directed therapy may be broadly applicable across diverse subtypes of NSCLC.
Collapse
Affiliation(s)
- Ross A Soo
- National University Hospital, Singapore, 119074, Singapore
| | | | | | - Chuntao Wu
- Daiichi Sankyo, Basking Ridge, NJ07920, USA
| | | | | | - Lukas Bubendorf
- Institute of Medical Genetics & Pathology, University Hospital Basel, 4031, Basel, Switzerland
| |
Collapse
|
9
|
Hassanzadeh Makoui R, Fekri S, Ansari N, Hassanzadeh Makoui M. Investigating the Co-Expression Rate of HER2 and HER3 Biomarkers in Cancer Patients: A Systematic Review and Meta-Analysis. Asian Pac J Cancer Prev 2024; 25:2979-2990. [PMID: 39342574 DOI: 10.31557/apjcp.2024.25.9.2979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Many types of cancer express the HER2/HER3 heterodimer, which is a crucial oncogenic unit. Research has shown that when these two biomarkers are expressed together, it correlates with higher tumor aggressiveness and lower overall survival rate. Therefore, many therapies have been developed to target both biomarkers simultaneously. This study aims to collect data on the co-expression levels of these biomarkers across different types of cancers. METHODS A comprehensive search was conducted across PubMed, Scopus, Embase, and Web of Science databases to identify relevant studies. The event rates and their corresponding 95% confidence intervals were calculated. Heterogeneity, subgroup, and meta-regression analyses were conducted based on patients' residency region, age, and gender. The protocol of this study was registered in PROSPERO under ID: CRD42024504256. RESULTS We have detected 60 studies that met all of the inclusion criteria for our research. Out of these, we have focused on a total of 19 studies (with 6,079 participants) related to breast cancer, 9 studies (with 829 participants) related to lung cancer, 6 studies (with 1423 participants) related to gastric cancer, and 4 studies (with 802 participants) related to colorectal cancer for conducting our meta-analysis. According to our results, the co-expression rate of HER2 and HER3 in breast cancer patients is 18.5% (95%CI 11.7-27.9), in colorectal cancer patients is 17.1% (95%CI 2.4-63.4), in gastric cancer patients is 11.3% (95%CI 4.2-17.2), and in lung cancer patients is 12.7% (95%CI 5.2-22.8). The co-expression of HER2 and HER3 in lung cancer has a significant association with patients' gender (P=0.038). CONCLUSION The study found that HER2 and HER3 biomarkers, which are targets for different therapies, are co-expressed in various types of cancer.
Collapse
Affiliation(s)
- Reza Hassanzadeh Makoui
- Department of Cardiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Shiva Fekri
- Department of Obstetrics and Gynecology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Negar Ansari
- Department of Internal Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | |
Collapse
|
10
|
Zhou L, Lu Y, Liu W, Wang S, Wang L, Zheng P, Zi G, Liu H, Liu W, Wei S. Drug conjugates for the treatment of lung cancer: from drug discovery to clinical practice. Exp Hematol Oncol 2024; 13:26. [PMID: 38429828 PMCID: PMC10908151 DOI: 10.1186/s40164-024-00493-8] [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: 01/06/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024] Open
Abstract
A drug conjugate consists of a cytotoxic drug bound via a linker to a targeted ligand, allowing the targeted delivery of the drug to one or more tumor sites. This approach simultaneously reduces drug toxicity and increases efficacy, with a powerful combination of efficient killing and precise targeting. Antibody‒drug conjugates (ADCs) are the best-known type of drug conjugate, combining the specificity of antibodies with the cytotoxicity of chemotherapeutic drugs to reduce adverse reactions by preferentially targeting the payload to the tumor. The structure of ADCs has also provided inspiration for the development of additional drug conjugates. In recent years, drug conjugates such as ADCs, peptide‒drug conjugates (PDCs) and radionuclide drug conjugates (RDCs) have been approved by the Food and Drug Administration (FDA). The scope and application of drug conjugates have been expanding, including combination therapy and precise drug delivery, and a variety of new conjugation technology concepts have emerged. Additionally, new conjugation technology-based drugs have been developed in industry. In addition to chemotherapy, targeted therapy and immunotherapy, drug conjugate therapy has undergone continuous development and made significant progress in treating lung cancer in recent years, offering a promising strategy for the treatment of this disease. In this review, we discuss recent advances in the use of drug conjugates for lung cancer treatment, including structure-based drug design, mechanisms of action, clinical trials, and side effects. Furthermore, challenges, potential approaches and future prospects are presented.
Collapse
Affiliation(s)
- Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wei Liu
- Department of Geriatrics, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shanglong Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lingling Wang
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengdou Zheng
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guisha Zi
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030000, China.
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030000, China.
| |
Collapse
|
11
|
Kim M, Ju HM, Song JY, Sampson J, Bayliss R, Choi J. HER3 overexpression: a predictive marker for poor prognosis in advanced ALK-positive non-small cell lung cancer treated with ALK inhibitors. Transl Lung Cancer Res 2024; 13:321-333. [PMID: 38496685 PMCID: PMC10938092 DOI: 10.21037/tlcr-23-804] [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: 12/03/2023] [Accepted: 02/02/2024] [Indexed: 03/19/2024]
Abstract
Background Anaplastic lymphoma kinase (ALK)-targeted tyrosine kinase inhibitors (TKIs) improve patient survival; however, some patients develop ALK-TKI resistance with unidentified mechanisms. We investigated ErbB family and c-MET expression in patients with ALK-positive non-small cell lung cancer (NSCLC) to understand their roles in the ALK-TKI response. Methods We studied 72 patients with advanced ALK-positive NSCLC with EML4-ALK fusion variant subtyping and immunostaining for c-MET, EGFR, HER2, and HER3 on tissue specimens both pre- (primary) and post-treatment (secondary) with ALK-TKI. We investigated the association of their expression with survival outcomes and assessed the effectiveness of combining ALK and EGFR inhibitors in ALK-positive NSCLC cell lines stimulated with the HER3-specific ligand HRG1. Results High expression of c-MET, EGFR, HER2, and HER3 was observed in 4.9%, 18.0%, 1.6%, and 25.8% of primary tumors, respectively, and 18.5%, 37.0%, 10.7%, and 35.7% of secondary tumors, respectively. HER3 overexpression in primary tumors showed inferior survival (P=0.132). In the subgroup with EML4-ALK variant 1/2 (V1/V2), HER3 overexpression was significantly associated with inferior survival in both primary and secondary tumors (P=0.022 and P=0.004, respectively). Combination treatment with lorlatinib and erlotinib significantly reduced HRG1-induced activation of RTK signaling in ALK-positive NSCLC cells. Conclusions HER3 overexpression has potential as a prognostic marker in ALK-positive NSCLCs, including ALK-TKI naïve and treated cases, especially those with EML4-ALK V1/V2. Assessing HER3 expression may be crucial for treatment planning and outcome prediction in these patients.
Collapse
Affiliation(s)
- Meejeong Kim
- Department of Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyun-min Ju
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji-young Song
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Josephina Sampson
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Richard Bayliss
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Jene Choi
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
12
|
Hachey SJ, Hatch CJ, Gaebler D, Mocherla A, Nee K, Kessenbrock K, Hughes CCW. Targeting tumor-stromal interactions in triple-negative breast cancer using a human vascularized micro-tumor model. Breast Cancer Res 2024; 26:5. [PMID: 38183074 PMCID: PMC10768273 DOI: 10.1186/s13058-023-01760-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 12/21/2023] [Indexed: 01/07/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive with limited available treatments. Stromal cells in the tumor microenvironment (TME) are crucial in TNBC progression; however, understanding the molecular basis of stromal cell activation and tumor-stromal crosstalk in TNBC is limited. To investigate therapeutic targets in the TNBC stromal niche, we used an advanced human in vitro microphysiological system called the vascularized micro-tumor (VMT). Using single-cell RNA sequencing, we revealed that normal breast tissue stromal cells activate neoplastic signaling pathways in the TNBC TME. By comparing interactions in VMTs with clinical data, we identified therapeutic targets at the tumor-stromal interface with potential clinical significance. Combining treatments targeting Tie2 signaling with paclitaxel resulted in vessel normalization and increased efficacy of paclitaxel in the TNBC VMT. Dual inhibition of HER3 and Akt also showed efficacy against TNBC. These data demonstrate the potential of inducing a favorable TME as a targeted therapeutic approach in TNBC.
Collapse
Affiliation(s)
- Stephanie J Hachey
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA.
| | | | - Daniela Gaebler
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Aneela Mocherla
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Kevin Nee
- Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Kai Kessenbrock
- Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Christopher C W Hughes
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
13
|
Chen Q, Jia G, Zhang X, Ma W. Targeting HER3 to overcome EGFR TKI resistance in NSCLC. Front Immunol 2024; 14:1332057. [PMID: 38239350 PMCID: PMC10794487 DOI: 10.3389/fimmu.2023.1332057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/13/2023] [Indexed: 01/22/2024] Open
Abstract
Receptor tyrosine kinases (RTKs) play a crucial role in cellular signaling and oncogenic progression. Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKIs) have become the standard treatment for advanced non-small cell lung cancer (NSCLC) patients with EGFR-sensitizing mutations, but resistance frequently emerges between 10 to 14 months. A significant factor in this resistance is the role of human EGFR 3 (HER3), an EGFR family member. Despite its significance, effective targeting of HER3 is still developing. This review aims to bridge this gap by deeply examining HER3's pivotal contribution to EGFR TKI resistance and spotlighting emerging HER3-centered therapeutic avenues, including monoclonal antibodies (mAbs), TKIs, and antibody-drug conjugates (ADCs). Preliminary results indicate combining HER3-specific treatments with EGFR TKIs enhances antitumor effects, leading to an increased objective response rate (ORR) and prolonged overall survival (OS) in resistant cases. Embracing HER3-targeting therapies represents a transformative approach against EGFR TKI resistance and emphasizes the importance of further research to optimize patient stratification and understand resistance mechanisms.
Collapse
Affiliation(s)
- Qiuqiang Chen
- Key Laboratory for Translational Medicine, The First Affiliated Hospital, Huzhou University, Huzhou, Zhejiang, China
| | - Gang Jia
- Department of Medical Oncology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xilin Zhang
- Key Laboratory for Translational Medicine, The First Affiliated Hospital, Huzhou University, Huzhou, Zhejiang, China
| | - Wenxue Ma
- Department of Medicine, Moores Cancer Center, and Sanford Stem Cell Institute, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
14
|
Ghosh I, Dey Ghosh R, Mukhopadhyay S. Identification of genes associated with gall bladder cell carcinogenesis: Implications in targeted therapy of gall bladder cancer. World J Gastrointest Oncol 2023; 15:2053-2063. [PMID: 38173427 PMCID: PMC10758643 DOI: 10.4251/wjgo.v15.i12.2053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/11/2023] [Accepted: 11/10/2023] [Indexed: 12/14/2023] Open
Abstract
Gall bladder cancer (GBC) is becoming a very devastating form of hepatobiliary cancer in India. Every year new cases of GBC are quite high in India. Despite recent advanced multimodality treatment options, the survival of GBC patients is very low. If the disease is diagnosed at the advanced stage (with local nodal metastasis or distant metastasis) or surgical resection is inoperable, the prognosis of those patients is very poor. So, perspectives of targeted therapy are being taken. Targeted therapy includes hormone therapy, proteasome inhibitors, signal transduction and apoptosis inhibitors, angiogenesis inhibitors, and immunotherapeutic agents. One such signal transduction inhibitor is the specific short interfering RNA (siRNA) or short hairpin RNA (shRNA). For developing siRNA-mediated therapy shRNA, although several preclinical studies to evaluate the efficacy of these key molecules have been performed using gall bladder cells, many more clinical trials are required. To date, many such genes have been identified. This review will discuss the recently identified genes associated with GBC and those that have implications in its treatment by siRNA or shRNA.
Collapse
Affiliation(s)
- Ishita Ghosh
- Department of Molecular Biology, Netaji Subhas Chandra Bose Cancer Research Institute, Kolkata 700094, India
| | - Ruma Dey Ghosh
- Department of Molecular Biology, Netaji Subhas Chandra Bose Cancer Research Institute, Kolkata 700094, India
| | - Soma Mukhopadhyay
- Department of Molecular Biology, Netaji Subhas Chandra Bose Cancer Research Institute, Kolkata 700094, India
| |
Collapse
|
15
|
Capone E, Tryggvason T, Cela I, Dufrusine B, Pinti M, Del Pizzo F, Gunnarsdottir HS, Grottola T, De Laurenzi V, Iacobelli S, Lattanzio R, Sala G. HER-3 surface expression increases in advanced colorectal cancer representing a potential therapeutic target. Cell Death Discov 2023; 9:400. [PMID: 37898642 PMCID: PMC10613198 DOI: 10.1038/s41420-023-01692-8] [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: 08/13/2023] [Revised: 09/27/2023] [Accepted: 10/16/2023] [Indexed: 10/30/2023] Open
Abstract
HER-3 (also known as ErbB-3) is a human epidermal growth factor receptor tyrosine kinases family member, and its expression in CRC (colorectal cancer) tissues was previously associated with poor prognosis. In this study, HER-3 expression was analyzed by immunohistochemistry in two cohorts of early and advanced metastatic CRC patients. The first cohort included 180 patients diagnosed with CRC in absence of lymph nodes or distant metastases (Stage I and Stage II), while the second was obtained from 53 advanced metastatic CRC patients who developed synchronous (SM) and metachronous (MM) liver metastases. In the first early-stage CRC cohort, 86 out of 180 (47.8%) tumors showed membranous expression of HER-3, with a mean percentage of positive tumor cells of 25.7%; conversely, in advanced metastatic CRC primary tumors, HER-3 was detected in all specimens, with a mean percentage of positive tumor cells of 76.1%. Kaplan-Meier curves showed that in the advanced metastatic CRC group, patients with HER-3high tumors had a significantly lower Cancer-Specific Survival (CSS) rate compared to patients with HER-3low tumors (p = 0.021). Importantly, this worse CSS rate was observed only in the MM subgroup of patients with HER-3high tumors (p = 0.002). Multivariate analysis confirmed that high HER-3 expression represents a significant and strong risk factor for death in patients developing MM liver metastases (Hazard Ratio = 64.9; 95% Confidence Interval, 4.7-886.6; p = 0.002). In addition, using a specific anti-HER-3 antibody-drug conjugate, named EV20/MMAF, we showed that HER-3 + CRC cells can be efficiently targeted in vitro and in vivo. Overall, this study confirms that surface HER-3 is highly expressed in CRC and reveals that HER-3 expression increases in metastatic CRC patients compared to early stage. Importantly, the results suggest that HER-3 has a prognostic and therapeutic value in patients developing MM liver metastases.
Collapse
Affiliation(s)
- Emily Capone
- Department of Innovative Technologies in Medicine & Dentistry, University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Thordur Tryggvason
- Department of Pathology, Landspítali - The National University Hospital of Iceland, Reykjavik, Iceland
| | - Ilaria Cela
- Department of Innovative Technologies in Medicine & Dentistry, University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Beatrice Dufrusine
- Center for Advanced Studies and Technology (CAST), University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Morena Pinti
- Department of Medical, Oral and Biotechnological Sciences, University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Francesco Del Pizzo
- Center for Advanced Studies and Technology (CAST), University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | | | - Tommaso Grottola
- Surgical Oncology Unit, Casa di Cura Pierangeli, 65124, Pescara, Italy
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine & Dentistry, University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | | | - Rossano Lattanzio
- Department of Innovative Technologies in Medicine & Dentistry, University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Gianluca Sala
- Department of Innovative Technologies in Medicine & Dentistry, University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy.
- Center for Advanced Studies and Technology (CAST), University "Gabriele d'Annunzio" of Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
16
|
Sharma T, Zhang Y, Zigrossi A, Cravatt BF, Kastrati I. Dimethyl fumarate inhibits ZNF217 and can be beneficial in a subset of estrogen receptor positive breast cancers. Breast Cancer Res Treat 2023; 201:561-570. [PMID: 37477798 DOI: 10.1007/s10549-023-07037-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 07/05/2023] [Indexed: 07/22/2023]
Abstract
PURPOSE The oncogenic factor ZNF217 promotes aggressive estrogen receptor (ER)+breast cancer disease suggesting that its inhibition may be useful in the clinic. Unfortunately, no direct pharmacological inhibitor is available. Dimethyl fumarate (DMF) exhibits anti-breast cancer activities, in vitro and in pre-clinical in vivo models. Its therapeutic benefits stem from covalent modification of cellular thiols such as protein cysteines, but the full profile of molecular targets mediating its anti-breast cancer effects remains to be determined. METHODS ER+breast cancer cells were treated with DMF followed by cysteine-directed proteomics. Cells with modulated ZNF217 levels were used to probe the efficacy of DMF. RESULTS Covalent modification of ZNF217 by DMF identified by proteomics was confirmed by using a DMF-chemical probe. Inhibition of ZNF217's transcriptional activity by DMF was evident on reported ZNF217-target genes. ZNF217 as an oncogene has been shown to enhance stem-like properties, survival, proliferation, and invasion. Consistent with ZNF217 inhibition, DMF was more effective at blocking these ZNF217-driven phenotypes in cells with elevated ZNF217 expression. Furthermore, partial knockdown of ZNF217 led to a reduction in DMF's efficacy. DMF's in vivo activity was evaluated in a xenograft model of MCF-7 HER2 cells that have elevated expression of ZNF217 and DMF treatment resulted in significant inhibition of tumor growth. CONCLUSION These data indicate that DMF's anti-breast cancer activities in the ER+HER2+models, at least in part, are due to inhibition of ZNF217. DMF is identified as a new covalent inhibitor of ZNF217.
Collapse
Affiliation(s)
- Tanu Sharma
- The Department of Cancer Biology, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA
| | - Yuanjin Zhang
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Alexandra Zigrossi
- The Department of Cancer Biology, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA
| | - Benjamin F Cravatt
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Irida Kastrati
- The Department of Cancer Biology, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA.
| |
Collapse
|
17
|
Takeda T, Tsubaki M, Genno S, Tokunaga K, Tanaka R, Nishida S. HER3/Akt/mTOR pathway is a key therapeutic target for the reduction of triple‑negative breast cancer metastasis via the inhibition of CXCR4 expression. Int J Mol Med 2023; 52:80. [PMID: 37477145 PMCID: PMC10555474 DOI: 10.3892/ijmm.2023.5283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/20/2023] [Indexed: 07/22/2023] Open
Abstract
Triple‑negative breast cancer (TNBC), a highly metastatic subtype of breast cancer, and it has the worst prognosis among all subtypes of breast cancer. However, no effective systematic therapy is currently available for TNBC metastasis. Therefore, novel therapies targeting the key molecular mechanisms involved in TNBC metastasis are required. The present study examined whether the expression levels of human epidermal growth factor receptor 3 (HER3) were associated with the metastatic phenotype of TNBC, and evaluated the potential of HER3 as a therapeutic target in vitro and in vivo. A new highly metastatic 4T1 TNBC cell line, termed 4T1‑L8, was established. The protein expression levels in 4T1‑L8 cells were measured using luminex magnetic bead assays and western blot analysis. The HER3 expression levels and distant metastasis‑free survival (DMFS) in TNBC were analyzed using Kaplan‑Meier Plotter. Transwell migration and invasion assays were performed to detect migration and invasion. The anti‑metastatic effects were determined in an experimental mouse model of metastasis. The results revealed that the increased expression of the HER3/Akt/mTOR pathway was associated with a greater level of cell migration, invasion and metastasis of TNBC cells. In addition, it was found that high expression levels of HER3 were associated with a poor DMFS. The inhibition of the HER3/Akt/mammalian target of rapamycin (mTOR) pathway decreased the migration, invasion and metastasis of TNBC cells by decreasing the expression of C‑X‑C chemokine receptor type 4 (CXCR4). Furthermore, treatment of metastatic TNBC cells with everolimus inhibited their migration, invasion and metastasis by decreasing CXCR4 expression. Thus, targeting the HER3/Akt/mTOR pathway opens up a new avenue for the development of therapeutics against TNBC metastasis; in addition, everolimus may prove to be an effective therapeutic agent for the suppression of TNBC metastasis.
Collapse
Affiliation(s)
- Tomoya Takeda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Shuji Genno
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Kenta Tokunaga
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Remi Tanaka
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| |
Collapse
|
18
|
Steele TM, Tsamouri MM, Siddiqui S, Lucchesi CA, Vasilatis D, Mooso BA, Durbin-Johnson BP, Ma AH, Hejazi N, Parikh M, Mudryj M, Pan CX, Ghosh PM. Cisplatin-induced increase in heregulin 1 and its attenuation by the monoclonal ErbB3 antibody seribantumab in bladder cancer. Sci Rep 2023; 13:9617. [PMID: 37316561 PMCID: PMC10267166 DOI: 10.1038/s41598-023-36774-1] [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: 11/13/2022] [Accepted: 06/09/2023] [Indexed: 06/16/2023] Open
Abstract
Cisplatin-based combination chemotherapy is the foundation for treatment of advanced bladder cancer (BlCa), but many patients develop chemoresistance mediated by increased Akt and ERK phosphorylation. However, the mechanism by which cisplatin induces this increase has not been elucidated. Among six patient-derived xenograft (PDX) models of BlCa, we observed that the cisplatin-resistant BL0269 express high epidermal growth factor receptor, ErbB2/HER2 and ErbB3/HER3. Cisplatin treatment transiently increased phospho-ErbB3 (Y1328), phospho-ERK (T202/Y204) and phospho-Akt (S473), and analysis of radical cystectomy tissues from patients with BlCa showed correlation between ErbB3 and ERK phosphorylation, likely due to the activation of ERK via the ErbB3 pathway. In vitro analysis revealed a role for the ErbB3 ligand heregulin1-β1 (HRG1/NRG1), which is higher in chemoresistant lines compared to cisplatin-sensitive cells. Additionally, cisplatin treatment, both in PDX and cell models, increased HRG1 levels. The monoclonal antibody seribantumab, that obstructs ErbB3 ligand-binding, suppressed HRG1-induced ErbB3, Akt and ERK phosphorylation. Seribantumab also prevented tumor growth in both the chemosensitive BL0440 and chemoresistant BL0269 models. Our data demonstrate that cisplatin-associated increases in Akt and ERK phosphorylation is mediated by an elevation in HRG1, suggesting that inhibition of ErbB3 phosphorylation may be a useful therapeutic strategy in BlCa with high phospho-ErbB3 and HRG1 levels.
Collapse
Affiliation(s)
- Thomas M Steele
- Research Service, VA Northern California Health Care System, Mather, CA, USA
- Department of Urological Surgery, University of California Davis School of Medicine, 4860 Y Street, Suite 3500, Sacramento, CA, 95817, USA
| | - Maria Malvina Tsamouri
- Research Service, VA Northern California Health Care System, Mather, CA, USA
- Department of Urological Surgery, University of California Davis School of Medicine, 4860 Y Street, Suite 3500, Sacramento, CA, 95817, USA
| | - Salma Siddiqui
- Research Service, VA Northern California Health Care System, Mather, CA, USA
| | - Christopher A Lucchesi
- Research Service, VA Northern California Health Care System, Mather, CA, USA
- Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, USA
| | - Demitria Vasilatis
- Research Service, VA Northern California Health Care System, Mather, CA, USA
- Department of Urological Surgery, University of California Davis School of Medicine, 4860 Y Street, Suite 3500, Sacramento, CA, 95817, USA
| | - Benjamin A Mooso
- Research Service, VA Northern California Health Care System, Mather, CA, USA
| | - Blythe P Durbin-Johnson
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis, Davis, CA, USA
| | - Ai-Hong Ma
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Nazila Hejazi
- Research Service, VA Northern California Health Care System, Mather, CA, USA
- Yosemite Pathology Medical Group, Inc., Modesto, CA, USA
| | - Mamta Parikh
- Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis, Sacramento, CA, USA
| | - Maria Mudryj
- Research Service, VA Northern California Health Care System, Mather, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Chong-Xian Pan
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paramita M Ghosh
- Research Service, VA Northern California Health Care System, Mather, CA, USA.
- Department of Urological Surgery, University of California Davis School of Medicine, 4860 Y Street, Suite 3500, Sacramento, CA, 95817, USA.
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis, Davis, CA, USA.
| |
Collapse
|
19
|
Morato A, Accornero P, Hovey RC. ERBB Receptors and Their Ligands in the Developing Mammary Glands of Different Species: Fifteen Characters in Search of an Author. J Mammary Gland Biol Neoplasia 2023; 28:10. [PMID: 37219601 DOI: 10.1007/s10911-023-09538-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/26/2023] [Indexed: 05/24/2023] Open
Abstract
The ERBB tyrosine kinase receptors and their ligands belong to a complex family that has diverse biological effects and expression profiles in the developing mammary glands, where its members play an essential role in translating hormone signals into local effects. While our understanding of these processes stems mostly from mouse models, there is the potential for differences in how this family functions in the mammary glands of other species, particularly in light of their unique histomorphological features. Herein we review the postnatal distribution and function of ERBB receptors and their ligands in the mammary glands of rodents and humans, as well as for livestock and companion animals. Our analysis highlights the diverse biology for this family and its members across species, the regulation of their expression, and how their roles and functions might be modulated by varying stromal composition and hormone interactions. Given that ERBB receptors and their ligands have the potential to influence processes ranging from normal mammary development to diseased states such as cancer and/or mastitis, both in human and veterinary medicine, a more complete understanding of their biological functions should help to direct future research and the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Alessia Morato
- Department of Animal Science, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA.
| | - Paolo Accornero
- Department of Veterinary Science, University of Turin, Largo Paolo Braccini 2, Grugliasco, TO, 10095, Italy
| | - Russell C Hovey
- Department of Animal Science, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| |
Collapse
|
20
|
Desai O, Wang R. HER3- A key survival pathway and an emerging therapeutic target in metastatic colorectal cancer and pancreatic ductal adenocarcinoma. Oncotarget 2023; 14:439-443. [PMID: 37163206 PMCID: PMC10171365 DOI: 10.18632/oncotarget.28421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Colorectal cancer (CRC) and pancreatic ductal adenocarcinoma (PDAC) are highly metastatic cancers with poor survival rates. The tumor microenvironment has been shown to play a critical role in cancer progression and response to therapies. Endothelial cells (ECs) are a key component of the tumor microenvironment and promote cancer cell survival by secreting soluble factors that activate cancer-promoting signaling pathways. Studies from us and others identified HER3 as a key mediator of liver EC-induced chemoresistance and cancer cell growth in metastatic CRC and PDAC. In this article, we discuss that HER3-targeted therapies may be effective in treating patients with HER3-expressing CRC and PDAC, and highlight the importance of applying HER3 expression as a predictive biomarker for patient response to HER3-targeted therapies. We also discuss the challenges encountered in past clinical trials of HER3-targeted therapies, including the role of NRG1 gene fusions, alternative HER3 activation mechanisms, and adaptive resistance mechanisms. Finally, we conclude by suggesting the future directions of HER3-targeted therapies, including novel approaches to overcome chemoresistance and promote cancer cell death.
Collapse
Affiliation(s)
- Omkar Desai
- Department of Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Rui Wang
- Department of Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
21
|
Deshmukh R, Prajapati M, Harwansh RK. A review on emerging targeted therapies for the management of metastatic colorectal cancers. Med Oncol 2023; 40:159. [PMID: 37097307 DOI: 10.1007/s12032-023-02020-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/08/2023] [Indexed: 04/26/2023]
Abstract
Colorectal cancers are among the most commonly found cancers over the world. In spite of the recent advancements in diagnosis and prognosis, the management of this metastatic condition remains a challenge. The utility of monoclonal antibodies in the healing of patients with colorectal cancer has opened a new chapter in the quest for newer therapies. The resistance to the standard treatment regimen made it mandatory to search for newer targets. Mutagenic alterations in the gene engaged in cellular differentiation and growth pathway have been the reason for resistance to treatment. The newer therapies target the various proteins and receptors involved in the signal transduction and down streaming pathways leading to cell proliferation. This review presents an insight into the newer targeted therapies for colorectal cancer involving tyrosine kinase blockers, epidermal growth factor receptors, vascular endothelial growth factor, immune checkpoint therapy, and BRAF inhibitors.
Collapse
Affiliation(s)
- Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India.
| | - Mahendra Prajapati
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| |
Collapse
|
22
|
Daoui O, Mali SN, Elkhattabi K, Elkhattabi S, Chtita S. Repositioning Cannabinoids and Terpenes as Novel EGFR-TKIs Candidates for Targeted Therapy Against Cancer: A virtual screening model using CADD and biophysical simulations. Heliyon 2023; 9:e15545. [PMID: 37128337 PMCID: PMC10148140 DOI: 10.1016/j.heliyon.2023.e15545] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 05/03/2023] Open
Abstract
This study examines the potential of Cannabis sativa L. plants to be repurposed as therapeutic agents for cancer treatment through designing of hybrid Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs). A set of 50 phytochemicals was taken from Cannabinoids and Terpenes and subjected for screening using Semi-flexible and Flexible Molecular Docking methods, MM-GBSA free binding energy computations, and pharmacokinetic/pharmacodynamic (ADME-Tox) predictions. Nine promising phytochemicals, Cannabidiolic acid (CBDA), Cannabidiol (CBD), Tetrahydrocannabivarin (THCV), Dronabinol (Δ-9-THC), Delta-8-Tetrahydrocannabinol (Δ-8-THC), Cannabicyclol (CBL), Delta9-tetrahydrocannabinolic acid (THCA), Beta-Caryophyllene (BCP), and Gamma-Elemene (γ-Ele) were identified as potential EGFR-TKIs natural product candidates for cancer therapy. To further validate these findings, a set of Molecular Dynamics simulations were conducted over a 200 ns trajectory. This hybrid early drug discovery screening strategy has the potential to yield a new generation of EGFR-TKIs based on natural cannabis products, suitable for cancer therapy. In addition, the application of this computational strategy in the virtual screening of both natural and synthetic chemical libraries could support the discovery of a wide range of lead drug agents to address numerous diseases.
Collapse
Affiliation(s)
- Ossama Daoui
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, P.O. Box 72, Fez, Morocco
- Corresponding author.
| | - Suraj N. Mali
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, India, 835215
| | - Kaouakeb Elkhattabi
- Department of Fundamental Sciences, Faculty of Dental Medicine, Mohammed V University in Rabat, Morocco
| | - Souad Elkhattabi
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, P.O. Box 72, Fez, Morocco
| | - Samir Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, P.O. Box 7955, Casablanca, Morocco
| |
Collapse
|
23
|
Rathore M, Zhang W, Wright M, Zarei M, Vaziri-Gohar A, Hajihassani O, Abbas A, Feng H, Brody J, Markowitz SD, Winter J, Wang R. Liver Endothelium Microenvironment Promotes HER3-mediated Cell Growth in Pancreatic Ductal Adenocarcinoma. JOURNAL OF CANCER SCIENCE AND CLINICAL THERAPEUTICS 2022; 6:431-445. [PMID: 36644317 PMCID: PMC9838560 DOI: 10.26502/jcsct.5079182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
~90% metastatic pancreatic ductal adenocarcinoma (mPDAC) occurs in the liver, and the 5-year survival rate for patients with mPDAC is only at 3%. The liver has a unique endothelial cell (EC)-rich microenvironment, and preclinical studies showed that ECs promote cancer cell survival pathways by secreting soluble factors in a paracrine fashion in other types of cancer. However, the effects of liver ECs on mPDAC have not been elucidated. In this study, we used primary liver ECs and determined that liver EC-secreted factors containing conditioned medium (CM) increased PDAC cell growth, compared to control CM from PDAC cells. Using an unbiased receptor tyrosine kinase array, we identified human epidermal growth factor receptor 3 (HER3, also known as ErbB3) as a key mediator of liver EC-induced growth in PDAC cells with HER3 expression (HER3 +ve). We found that EC-secreted neuregulins activated the HER3-AKT signaling axis, and that depleting neuregulins from EC CM or blocking HER3 with an antibody, seribantumab, attenuated EC-induced functions in HER3 +ve PDAC cells, but not in cells without HER3 expression. Furthermore, we determined that EC CM increased PDAC xenograft growth in vivo, and that seribantumab blocked EC-induced growth in xenografts with HER3 expression. These findings elucidated a paracrine role of liver ECs in promoting PDAC cell growth, and identified the HER3-AKT axis as a key mediator in EC-induced functions in HER3 +ve PDAC cells. As over 70% mPDAC express HER3, this study highlights the potential of using HER3-targeted therapies for treating patients with HER3 +ve mPDAC.
Collapse
Affiliation(s)
- Moeez Rathore
- Department of Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
| | - Wei Zhang
- Department of Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
| | - Michel'le Wright
- Department of Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
| | - Mehrdad Zarei
- Department of Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
| | - Ali Vaziri-Gohar
- Department of Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
| | - Omid Hajihassani
- Department of Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
| | - Ata Abbas
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
| | - Hao Feng
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jonathan Brody
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239
| | - Sanford D Markowitz
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
- Department of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106. USA
| | - Jordan Winter
- Department of Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland, OH 44106
| | - Rui Wang
- Department of Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland, OH 44106
| |
Collapse
|
24
|
Nussinov R, Tsai CJ, Jang H. A New View of Activating Mutations in Cancer. Cancer Res 2022; 82:4114-4123. [PMID: 36069825 PMCID: PMC9664134 DOI: 10.1158/0008-5472.can-22-2125] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/16/2022] [Accepted: 09/01/2022] [Indexed: 12/14/2022]
Abstract
A vast effort has been invested in the identification of driver mutations of cancer. However, recent studies and observations call into question whether the activating mutations or the signal strength are the major determinant of tumor development. The data argue that signal strength determines cell fate, not the mutation that initiated it. In addition to activating mutations, factors that can impact signaling strength include (i) homeostatic mechanisms that can block or enhance the signal, (ii) the types and locations of additional mutations, and (iii) the expression levels of specific isoforms of genes and regulators of proteins in the pathway. Because signal levels are largely decided by chromatin structure, they vary across cell types, states, and time windows. A strong activating mutation can be restricted by low expression, whereas a weaker mutation can be strengthened by high expression. Strong signals can be associated with cell proliferation, but too strong a signal may result in oncogene-induced senescence. Beyond cancer, moderate signal strength in embryonic neural cells may be associated with neurodevelopmental disorders, and moderate signals in aging may be associated with neurodegenerative diseases, like Alzheimer's disease. The challenge for improving patient outcomes therefore lies in determining signaling thresholds and predicting signal strength.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, NCI, Frederick, Maryland
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, NCI, Frederick, Maryland
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, NCI, Frederick, Maryland
| |
Collapse
|
25
|
Al taweraqi N, King RD. Improved prediction of gene expression through integrating cell signalling models with machine learning. BMC Bioinformatics 2022; 23:323. [PMID: 35933367 PMCID: PMC9356471 DOI: 10.1186/s12859-022-04787-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/13/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND A key problem in bioinformatics is that of predicting gene expression levels. There are two broad approaches: use of mechanistic models that aim to directly simulate the underlying biology, and use of machine learning (ML) to empirically predict expression levels from descriptors of the experiments. There are advantages and disadvantages to both approaches: mechanistic models more directly reflect the underlying biological causation, but do not directly utilize the available empirical data; while ML methods do not fully utilize existing biological knowledge. RESULTS Here, we investigate overcoming these disadvantages by integrating mechanistic cell signalling models with ML. Our approach to integration is to augment ML with similarity features (attributes) computed from cell signalling models. Seven sets of different similarity feature were generated using graph theory. Each set of features was in turn used to learn multi-target regression models. All the features have significantly improved accuracy over the baseline model - without the similarity features. Finally, the seven multi-target regression models were stacked together to form an overall prediction model that was significantly better than the baseline on 95% of genes on an independent test set. The similarity features enable this stacking model to provide interpretable knowledge about cancer, e.g. the role of ERBB3 in the MCF7 breast cancer cell line. CONCLUSION Integrating mechanistic models as graphs helps to both improve the predictive results of machine learning models, and to provide biological knowledge about genes that can help in building state-of-the-art mechanistic models.
Collapse
Affiliation(s)
- Nada Al taweraqi
- Department of Computer Science, University of Manchester, Manchester, UK
- Department of Computer Science, Taif University, Taif, Saudi Arabia
| | - Ross D. King
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Alan Turing Institute, London, UK
| |
Collapse
|
26
|
Barakat A, Abu-Serie MM, Ali M, Al-Majid AM, Ashraf S, Zia K, Ul-Haq Z, Al-Dhfyan A, Abdel-Aziz HA, El-Faham A, Domingo LR. Synthesis, In Vitro and in Cell Study of a New Spirooxindoles-Based N-Alkylated Maleimides Targeting HER2/3 Signaling Pathway. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2101486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Assem Barakat
- Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Marwa M. Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El Arab, Egypt
| | - M. Ali
- Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | | | - Sajda Ashraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Komal Zia
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Abdullah Al-Dhfyan
- Stem Cell Therapy and Tissue Re-Engineering Program, King Faisal Specialized Hospital and Research Center, Riyadh, Saudi Arabia
| | - Hatem A. Abdel-Aziz
- Department of Applied Organic Chemistry, National Research Center, Giza, Egypt
| | - Ayman El-Faham
- Department of Chemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Luis R. Domingo
- Department of Organic Chemistry, University of Valencia, Valencia, Spain
| |
Collapse
|
27
|
EGFR, HER2, and HER3 protein expression in paired primary tumor and lymph node metastasis of colorectal cancer. Sci Rep 2022; 12:12894. [PMID: 35902718 PMCID: PMC9334602 DOI: 10.1038/s41598-022-17210-2] [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: 01/24/2022] [Accepted: 07/21/2022] [Indexed: 12/24/2022] Open
Abstract
Due to the difficulty in sampling of metastatic tumors, patient selection is commonly based on results of primary tumor samples when metastatic samples are not available. However, due to tumor heterogeneity, metastatic tumors may be different from primary tumors in their phenotypes. The aim of this study was to investigate the expression of EGFR, HER2, and HER3 between primary and lymph node metastatic lesions of colorectal cancer. Paired primary tumors and lymph node metastases from 79 patients with colorectal cancer were retrospectively collected and analyzed for EGFR, HER2, and HER3 expression. High EGFR, HER2, and HER3 expression (2+ and 3+) was found in 64.2%, 66.0%, and 85.0% of primary tumors, and 56.8%, 46.0%, and 76.0% of lymph node metastases, respectively. Correlation rates between primary and metastatic lesions were 67.1%, 63.3%, and 74.7% for EGFR, HER2, and HER3, respectively. Stage IV tumors (with distant metastasis) had higher correlation rates of HER2 expression compared to stage III tumors (without distant metastasis) (P = 0.050). Moderate correlation rates in EGFR, HER2, and HER3 expression were observed between primary and metastatic lesions of colorectal cancer. Tumor stage or existence of distant metastasis could serve as potential predictive markers for the correlation of HER2 expression between primary tumors and lymph node metastases of colorectal cancer.
Collapse
|
28
|
朱 以, 王 志. [Current Progress and Future Developments of Antibody Drug Conjugates
in Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:468-476. [PMID: 35899443 PMCID: PMC9346152 DOI: 10.3779/j.issn.1009-3419.2022.102.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 11/20/2022]
Abstract
Antibody drug conjugates (ADCs) are a novel class of anti-cancer drugs, which combined the specificity of monoclonal antibodies with the cytotoxic palyload via the linkers. Many ADCs have not only verified impressive activity in a variety of cancers, including breast cancer and hematological system tumors, but also in lung cancer. The aim of this study was to provide informations for practice by summarizing the mechanism of action, clinical application and problems and challenges of ADCs.
.
Collapse
Affiliation(s)
- 以香 朱
- />100021 北京,国家癌症中心,国家肿瘤临床医学研究中心,中国医学科学院北京协和医学院肿瘤医院内科CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing 100021, China
| | - 志杰 王
- />100021 北京,国家癌症中心,国家肿瘤临床医学研究中心,中国医学科学院北京协和医学院肿瘤医院内科CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
29
|
Targeting Tumor Cells Overexpressing the Human Epidermal Growth Factor Receptor 3 with Potent Drug Conjugates Based on Affibody Molecules. Biomedicines 2022; 10:biomedicines10061293. [PMID: 35740315 PMCID: PMC9219639 DOI: 10.3390/biomedicines10061293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/22/2022] Open
Abstract
Increasing evidence suggests that therapy targeting the human epidermal growth factor receptor 3 (HER3) could be a viable route for targeted cancer therapy. Here, we studied a novel drug conjugate, ZHER3-ABD-mcDM1, consisting of a HER3-targeting affibody molecule, coupled to the cytotoxic tubulin polymerization inhibitor DM1, and an albumin-binding domain for in vivo half-life extension. ZHER3-ABD-mcDM1 showed a strong affinity to the extracellular domain of HER3 (KD 6 nM), and an even stronger affinity (KD 0.2 nM) to the HER3-overexpressing pancreatic carcinoma cell line, BxPC-3. The drug conjugate showed a potent cytotoxic effect on BxPC-3 cells with an IC50 value of 7 nM. Evaluation of a radiolabeled version, [99mTc]Tc-ZHER3-ABD-mcDM1, showed a relatively high rate of internalization, with a 27% internalized fraction after 8 h. Further in vivo evaluation showed that it could target BxPC-3 (pancreatic carcinoma) and DU145 (prostate carcinoma) xenografts in mice, with an uptake peaking at 6.3 ± 0.4% IA/g at 6 h post-injection for the BxPC-3 xenografts. The general biodistribution showed uptake in the liver, lung, salivary gland, stomach, and small intestine, organs known to express murine ErbB3 naturally. The results from the study show that ZHER3-ABD-mcDM1 is a highly potent and selective drug conjugate with the ability to specifically target HER3 overexpressing cells. Further pre-clinical and clinical development is discussed.
Collapse
|
30
|
HER2-CDH1 Interaction via Wnt/B-Catenin Is Associated with Patients' Survival in HER2-Positive Metastatic Gastric Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14051266. [PMID: 35267574 PMCID: PMC8909509 DOI: 10.3390/cancers14051266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary A deeper understanding of the molecular mechanisms involved in gastric cacner (GC) pathologenesis would help the identification of prognostic biomarkers and the development of new treatments. Human epidermal growth factor receptor 2 (HER2/ErbB2), a membrane-bound receptor of the EGFR family, may be overexpressed in GC. Trastuzumab is a HER2 inhibitor used to treat HER2+ metastatic gastric cancer (mGC). The present study aims to investigate the relationship between CDH1 mRNA expression and HER2-positivity in mGC using a multiplexed gene expression profile in two series of GC patients: 38 HER2+ and HER2- mGC and 36 HER2- GC with and without metastasis. Our results revealed the relationship between CDH1 and HER2 mRNA expression in mGC via the canonical WNT/β-catenin pathway and identified EGF as an independent prognostic biomarker for survival. Abstract Trastuzumab is a human epidermal growth factor receptor 2 (HER2) inhibitor used to treat HER2+ metastatic gastric cancer (mGC). The present study aims to investigate the relationship between CDH1 mRNA expression and HER2-positivity in mGC using a multiplexed gene expression profile in two series of gastric cancer (GC): Series 1 (n = 38): HER2+ and HER2- mGC; Series 2 (n = 36) HER2- GC with and without metastasis. To confirm the results, the same expression profiles were analyzed in 354 GC from The Cancer Genome Atlas (TCGA) dataset. The difference in gene expression connected HER2 overexpression with canonical wingless-type (Wnt)/β-catenin pathway and immunohistochemical (IHC) expression loss of E-cadherin (E-CAD). CDH1 mRNA expression was simultaneously associated with the rs16260-A variant and an increase in E-CAD expression. Differences in retinoic acid receptor alfa (RARA), RPL19 (coding for the 60S ribosomal L19 protein), catenin delta 1 (CTNND1), and epidermal growth factor (EGF) mRNA levels—all included in the Wnt/β-catenin pathway—were found associated with overall survival (OS). RARA, CTNND1, and EGF resulted in independent OS prognostic factors. EGF was confirmed as an independent factor along with TNM stage in HER2-overpressed mGC from TCGA collection. Our study highlighted factors involved in the WNT/β-catenin pathway that interconnected E-CAD with HER2 overexpression and patient survival.
Collapse
|
31
|
Hassani D, Jeddi-Tehrani M, Yousefi P, Mansouri-Fard S, Mobini M, Ahmadi-Zare H, Golsaz-Shirazi F, Amiri MM, Shokri F. Differential tumor inhibitory effects induced by HER3 extracellular subdomain-specific mouse monoclonal antibodies. Cancer Chemother Pharmacol 2022; 89:347-361. [PMID: 35079876 DOI: 10.1007/s00280-021-04390-3] [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: 09/01/2021] [Accepted: 12/15/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE The therapeutic potential of targeting the human epidermal growth factor receptor-3 (ErbB3/HER3) has long been ignored due to impaired tyrosine kinase function and low expression level in tumor cells compared with EGFR and HER2. Although recent investigations have explored the potential benefit of HER3 targeting and several anti-HER3 agents have been developed, there is still a critical need to design and produce more efficient therapeutics. This study was designed to develop tumor inhibitory monoclonal antibodies (MAbs) against different extracellular subdomains of HER3. METHODS Distinct extracellular subdomains of HER3 (DI+II and DIII+IV) were utilized to produce MAbs by hybridoma technology. Biochemical and functional characteristics of these MAbs were then investigated by various methodologies, including immunoblotting, flow cytometry, cell proliferation, cell signaling, and enzyme-linked immunosorbent assays. RESULTS Four anti-DI+II and six anti-DIII+IV MAbs were obtained, selected based on their ability to bind recombinant full HER3 extracellular domain (ECD). Our data showed that only one anti-DI+II and four anti-DIII+IV MAbs recognized the native form of HER3 by immunoblotting. Four MAbs recognized the membranous HER3 by flow cytometry leading to induction of different levels of receptor internalization and subsequent degradation. Results of cell proliferation assays using these MAbs indicated that they differentially inhibited proliferation of HER3-expressing cancer cells and showed considerable synergistic effects in combination with trastuzumab. Selected MAb with the highest inhibitory effect significantly inhibited the phosphorylation of AKT and ERK1/2 molecules. CONCLUSION Some of the anti-HER3 MAbs produced in this study displayed tumor inhibitory function and may be considered promising candidates for future HER3-targeted cancer therapy.
Collapse
Affiliation(s)
- Danesh Hassani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Parisa Yousefi
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Samaneh Mansouri-Fard
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mobini
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Hengameh Ahmadi-Zare
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Forough Golsaz-Shirazi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Fazel Shokri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
EGFR and HER2 exon 20 insertions in solid tumours: from biology to treatment. Nat Rev Clin Oncol 2022; 19:51-69. [PMID: 34561632 DOI: 10.1038/s41571-021-00558-1] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 02/08/2023]
Abstract
Protein tyrosine kinases of the human epidermal growth factor receptor family, including EGFR and HER2, have emerged as important therapeutic targets in non-small-cell lung, breast and gastroesophageal cancers, and are of relevance for the treatment of various other malignancies (particularly colorectal cancer). Classic activating EGFR exon 19 deletions and exon 21 mutations, and HER2 amplification and/or overexpression, are predictive of response to matched molecularly targeted therapies, translating into favourable objective response rates and survival outcomes. By comparison, cancers with insertion mutations in exon 20 of either EGFR or HER2 are considerably less sensitive to the currently available tyrosine kinase inhibitors and antibodies targeting these receptors. These exon 20 insertions are structurally distinct from other EGFR and HER2 mutations, providing an explanation for this lack of sensitivity. In this Review, we first discuss the prevalence and pan-cancer distribution of EGFR and HER2 exon 20 insertions, their biology and detection, and associated responses to current molecularly targeted therapies and immunotherapies. We then focus on novel approaches that are being developed to more effectively target tumours driven by these non-classic EGFR and HER2 alterations.
Collapse
|
33
|
Xie J, Zou Y, Gao T, Xie L, Tan D, Xie X. Therapeutic Landscape of Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer. Cancer Control 2022; 29:10732748221099230. [PMID: 35499382 PMCID: PMC9067050 DOI: 10.1177/10732748221099230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/30/2022] [Accepted: 04/18/2022] [Indexed: 01/08/2023] Open
Abstract
Human epidermal growth factor receptor 2-positive breast cancer (HER2+BC) is a common malignancy that is prone to recurrence and metastasis in the early stages, resulting in a poor prognosis for patients. Many studies have suggested that targeted therapy promotes clinical outcomes in HER2+BC. With the introduction of trastuzumab in 1998, the prognosis of patients with early HER2+BC has improved significantly. However, owing to obstinate drug resistance and adverse events, the addition of new agents in standardized treatment has become a research hotspot. These promising agents include antibodies, antibody-drug conjugates, tyrosine kinase inhibitors, and anti-HER2 combined therapies. This article provides a brief description of the biology of BC and the expression of HER2, with the aim to provide an overview of the therapeutic landscape of HER2+BC by reviewing research results and introducing the latest evidence to provide a reference for clinical treatment.
Collapse
Affiliation(s)
- Jindong Xie
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yutian Zou
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ting Gao
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Liming Xie
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Duxun Tan
- South China Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Xiaoming Xie
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
34
|
Zachariah NN, Basu A, Gautam N, Ramamoorthi G, Kodumudi KN, Kumar NB, Loftus L, Czerniecki BJ. Intercepting Premalignant, Preinvasive Breast Lesions Through Vaccination. Front Immunol 2021; 12:786286. [PMID: 34899753 PMCID: PMC8652247 DOI: 10.3389/fimmu.2021.786286] [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: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) prevention remains the ultimate cost-effective method to reduce the global burden of invasive breast cancer (IBC). To date, surgery and chemoprevention remain the main risk-reducing modalities for those with hereditary cancer syndromes, as well as high-risk non-hereditary breast lesions such as ADH, ALH, or LCIS. Ductal carcinoma in situ (DCIS) is a preinvasive malignant lesion of the breast that closely mirrors IBC and, if left untreated, develops into IBC in up to 50% of lesions. Certain high-risk patients with DCIS may have a 25% risk of developing recurrent DCIS or IBC, even after surgical resection. The development of breast cancer elicits a strong immune response, which brings to prominence the numerous advantages associated with immune-based cancer prevention over drug-based chemoprevention, supported by the success of dendritic cell vaccines targeting HER2-expressing BC. Vaccination against BC to prevent or interrupt the process of BC development remains elusive but is a viable option. Vaccination to intercept preinvasive or premalignant breast conditions may be possible by interrupting the expression pattern of various oncodrivers. Growth factors may also function as potential immune targets to prevent breast cancer progression. Furthermore, neoantigens also serve as effective targets for interception by virtue of strong immunogenicity. It is noteworthy that the immune response also needs to be strong enough to result in target lesion elimination to avoid immunoediting as it may occur in IBC arising from DCIS. Overall, if the issue of vaccine targets can be solved by interrupting premalignant lesions, there is a potential to prevent the development of IBC.
Collapse
Affiliation(s)
| | - Amrita Basu
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Namrata Gautam
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Ganesan Ramamoorthi
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Krithika N Kodumudi
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Nagi B Kumar
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Loretta Loftus
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Brian J Czerniecki
- Department of Breast Surgery, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
35
|
Kim K, Lee D. ERBB3-dependent AKT and ERK pathways are essential for atrioventricular cushion development in mouse embryos. PLoS One 2021; 16:e0259426. [PMID: 34714866 PMCID: PMC8555822 DOI: 10.1371/journal.pone.0259426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 10/19/2021] [Indexed: 11/19/2022] Open
Abstract
ERBB family members and their ligands play an essential role in embryonic heart development and adult heart physiology. Among them, ERBB3 is a binding partner of ERBB2; the ERBB2/3 complex mediates downstream signaling for cell proliferation. ERBB3 has seven consensus binding sites to the p85 regulatory subunit of PI3K, which activates the downstream AKT pathway, leading to the proliferation of various cells. This study generated a human ERBB3 knock-in mouse expressing a mutant ERBB3 whose seven YXXM p85 binding sites were replaced with YXXA. Erbb3 knock-in embryos exhibited lethality between E12.5 to E13.5, and showed a decrease in mesenchymal cell numbers and density in AV cushions. We determined that the proliferation of mesenchymal cells in the atrioventricular (AV) cushion in Erbb3 knock-in mutant embryos was temporarily reduced due to the decrease of AKT and ERK1/2 phosphorylation. Overall, our results suggest that AKT/ERK activation by the ERBB3-dependent PI3K signaling is crucial for AV cushion morphogenesis during embryonic heart development.
Collapse
Affiliation(s)
- Kyoungmi Kim
- Department of Physiology and Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
- * E-mail: (KK); (DL)
| | - Daekee Lee
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
- * E-mail: (KK); (DL)
| |
Collapse
|
36
|
HER3 PET Imaging: 68Ga-Labeled Affibody Molecules Provide Superior HER3 Contrast to 89Zr-Labeled Antibody and Antibody-Fragment-Based Tracers. Cancers (Basel) 2021; 13:cancers13194791. [PMID: 34638277 PMCID: PMC8508546 DOI: 10.3390/cancers13194791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary HER3 is a known driver for oncogenesis and therapy resistance in solid cancers. PET imaging could be a useful tool to non-invasively detect and monitor HER3 expression and aid in the selection of patients for HER3-targeted therapy. PET tracers based on therapeutic antibodies have thus far shown limited success in reliably imaging HER3-expressing tumors in clinical trials. Smaller-sized tracers specifically designed for imaging might be needed for higher contrast imaging and sufficient sensitivity. Our group has previously studied the use of radiolabeled affibody molecules for imaging of HER3 expression. In the present study, we compared four different types of potential PET tracers for imaging of HER3 expression in a preclinical model. We demonstrated that the affibody-based tracer, [68Ga]Ga-ZHER3, could provide overall superior imaging contrast to antibody- and antibody-fragment-based tracers shortly after injection. Our results indicate that HER3-targeting affibody molecules are promising agents for PET imaging of HER3 expression. Abstract HER3 (human epidermal growth factor receptor type 3) is a challenging target for diagnostic radionuclide molecular imaging due to the relatively modest overexpression in tumors and substantial expression in healthy organs. In this study, we compared four HER3-targeting PET tracers based on different types of targeting molecules in a preclinical model: the 89Zr-labeled therapeutic antibody seribantumab, a seribantumab-derived F(ab)2-fragment labeled with 89Zr and 68Ga, and the 68Ga-labeled affibody molecule [68Ga]Ga-ZHER3. The novel conjugates were radiolabeled and characterized in vitro using HER3-expressing BxPC-3 and DU145 human cancer cells. Biodistribution was studied using Balb/c nu/nu mice bearing BxPC-3 xenografts. HER3-negative RAMOS xenografts were used to demonstrate binding specificity in vivo. Autoradiography was conducted on the excised tumors. nanoPET/CT imaging was performed. New conjugates specifically bound to HER3 in vitro and in vivo. [68Ga]Ga-DFO-seribantumab-F(ab’)2 was considered unsuitable for imaging due to the low stability and high uptake in normal organs. The highest tumor-to-non-tumor contrast with [89Zr]Zr-DFO-seribantumab and [89Zr]Zr-DFO-seribantumab-F(ab’)2 was achieved at 96 h and 48 h pi, respectively. Despite lower tumor uptake, [68Ga]Ga-ZHER3 provided the best imaging contrast due to the fastest clearance from blood and normal organs. The results of our study suggest that affibody-based tracers are more suitable for PET imaging of HER3 expression than antibody- and antibody-fragment-based tracers.
Collapse
|
37
|
Eliseev IE, Ukrainskaya VM, Yudenko AN, Mikushina AD, Shmakov SV, Afremova AI, Ekimova VM, Vronskaia AA, Knyazev NA, Shamova OV. Targeting ErbB3 Receptor in Cancer with Inhibitory Antibodies from Llama. Biomedicines 2021; 9:biomedicines9091106. [PMID: 34572289 PMCID: PMC8467012 DOI: 10.3390/biomedicines9091106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 01/05/2023] Open
Abstract
The human ErbB3 receptor confers resistance to the pharmacological inhibition of EGFR and HER2 receptor tyrosine kinases in cancer, which makes it an important therapeutic target. Several anti-ErbB3 monoclonal antibodies that are currently being developed are all classical immunoglobulins. We took a different approach and discovered a group of novel heavy-chain antibodies targeting the extracellular domain of ErbB3 via a phage display of an antibody library from immunized llamas. We first produced three selected single-domain antibodies, named BCD090-P1, BCD090-M2, and BCD090-M456, in E. coli, as SUMO fusions that yielded up to 180 mg of recombinant protein per liter of culture. Then, we studied folding, aggregation, and disulfide bond formation, and showed their ultimate stability with half-denaturation of the strongest candidate, BCD090-P1, occurring in 8 M of urea. In surface plasmon resonance experiments, two most potent antibodies, BCD090-P1 and BCD090-M2, bound the extracellular domain of ErbB3 with 1.6 nM and 15 nM affinities for the monovalent interaction, respectively. The receptor binding was demonstrated by immunofluorescent confocal microscopy on four different ErbB3+ cancer cell lines. We observed that BCD090-P1 and BCD090-M2 bind noncompetitively to two distinct epitopes on the receptor. Both antibodies inhibited the ErbB3-driven proliferation of MCF-7 breast adenocarcinoma cells and HER2-overexpressing SK-BR-3 cells, with the EC50 in the range of 0.1–25 μg/mL. BCD090-M2 directly blocks ligand binding, whereas BCD090-P1 does not compete with the ligand and presumably acts through a distinct allosteric mechanism. We anticipate that these llama antibodies can be used to engineer new biparatopic anti-ErbB3 or bispecific anti-ErbB2/3 antibodies.
Collapse
Affiliation(s)
- Igor E. Eliseev
- Laboratory of Renewable Energy Sources, Alferov University, St. Petersburg 194021, Russia; (A.D.M.); (S.V.S.); (A.A.V.)
- Center for Personalized Medicine, FSBSI Institute of Experimental Medicine, St. Petersburg 197376, Russia;
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia;
- Correspondence:
| | - Valeria M. Ukrainskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia;
| | - Anna N. Yudenko
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny 141700, Russia;
| | - Anna D. Mikushina
- Laboratory of Renewable Energy Sources, Alferov University, St. Petersburg 194021, Russia; (A.D.M.); (S.V.S.); (A.A.V.)
| | - Stanislav V. Shmakov
- Laboratory of Renewable Energy Sources, Alferov University, St. Petersburg 194021, Russia; (A.D.M.); (S.V.S.); (A.A.V.)
| | | | | | - Anna A. Vronskaia
- Laboratory of Renewable Energy Sources, Alferov University, St. Petersburg 194021, Russia; (A.D.M.); (S.V.S.); (A.A.V.)
| | - Nickolay A. Knyazev
- Saint-Petersburg Clinical Scientific and Practical Center for Specialized Types of Medical Care (Oncological), St. Petersburg 197758, Russia;
| | - Olga V. Shamova
- Center for Personalized Medicine, FSBSI Institute of Experimental Medicine, St. Petersburg 197376, Russia;
| |
Collapse
|
38
|
Dan VM, Raveendran RS, Baby S. Resistance to Intervention: Paclitaxel in Breast Cancer. Mini Rev Med Chem 2021; 21:1237-1268. [PMID: 33319669 DOI: 10.2174/1389557520999201214234421] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/22/2020] [Accepted: 11/02/2020] [Indexed: 12/22/2022]
Abstract
Breast cancer stands as the most prevalent cancer in women globally, and contributes to the highest percentage of mortality due to cancer-related deaths in women. Paclitaxel (PTX) is heavily relied on as a frontline chemotherapy drug in breast cancer treatment, especially in advanced metastatic cancer. Generation of resistance to PTX often derails clinical management and adversely affects patient outcomes. Understanding the molecular mechanism of PTX resistance is necessary to device methods to aid in overcoming the resistance. Recent studies exploring the mechanism of development of PTX resistance have led to unveiling of a range novel therapeutic targets. PTX resistance pathways that involve major regulatory proteins/RNAs like RNF8/Twist/ROR1, TLR, ErbB3/ErbB2, BRCA1- IRIS, MENA, LIN9, MiRNA, FoxM1 and IRAK1 have expanded the complexity of resistance mechanisms, and brought newer insights into the development of drug targets. These resistance-related targets can be dealt with synthetic/natural therapeutics in combination with PTX. The present review encompasses the recent understanding of PTX resistance mechanisms in breast cancer and possible therapeutic combinations to overcome resistance.
Collapse
Affiliation(s)
- Vipin Mohan Dan
- Microbiology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Pacha-Palode 695562, Thiruvananthapuram, Kerala, India
| | - Reji Saradha Raveendran
- Microbiology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Pacha-Palode 695562, Thiruvananthapuram, Kerala, India
| | - Sabulal Baby
- Phytochemistry and Phytopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Pacha-Palode 695562, Thiruvananthapuram, Kerala, India
| |
Collapse
|
39
|
Barrett KE, Houson HA, Lin W, Lapi SE, Engle JW. Production, Purification, and Applications of a Potential Theranostic Pair: Cobalt-55 and Cobalt-58m. Diagnostics (Basel) 2021; 11:diagnostics11071235. [PMID: 34359318 PMCID: PMC8306844 DOI: 10.3390/diagnostics11071235] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 11/16/2022] Open
Abstract
The emerging success of [68Ga/177Lu]Ga/Lu-DOTATATE as a theranostic pair has spurred interest in other isotopes as potential theranostic combinations. Here, we review cobalt-55 and cobalt-58m as a potential theranostic pair. Radionuclidically pure cobalt-55 and cobalt-58m have been produced on small cyclotrons with high molar activity. In vitro, DOTATOC labeled with cobalt has shown greater affinity for SSTR2 than DOTATOC labeled with gallium and yttrium. Similarly, [58mCo]Co-DOTATATE has shown improved cell-killing capabilities as compared to DOTATATE labeled with either indium-111 or lutetium-177. Finally, PET imaging with an isotope such as cobalt-55 allows for image acquisition at much later timepoints than gallium, allowing for an increased degree of biological clearance of non-bound radiotracer. We discuss the accelerator targetry and radiochemistry used to produce cobalt-55,58m, emphasizing the implications of these techniques to downstream radiotracers being developed for imaging and therapy.
Collapse
Affiliation(s)
- Kendall E. Barrett
- Department of Medical Physics, University of Wisconsin, 1111 Highland Avenue, Madison, WI 53711, USA; (K.E.B.); (W.L.)
| | - Hailey A. Houson
- Department of Radiology, University of Alabama at Birmingham, 619 19th Street, Birmingham, AL 35294, USA; (H.A.H.); (S.E.L.)
| | - Wilson Lin
- Department of Medical Physics, University of Wisconsin, 1111 Highland Avenue, Madison, WI 53711, USA; (K.E.B.); (W.L.)
| | - Suzanne E. Lapi
- Department of Radiology, University of Alabama at Birmingham, 619 19th Street, Birmingham, AL 35294, USA; (H.A.H.); (S.E.L.)
| | - Jonathan W. Engle
- Department of Medical Physics, University of Wisconsin, 1111 Highland Avenue, Madison, WI 53711, USA; (K.E.B.); (W.L.)
- Department of Radiology, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792, USA
- Correspondence:
| |
Collapse
|
40
|
A scDb-based trivalent bispecific antibody for T-cell-mediated killing of HER3-expressing cancer cells. Sci Rep 2021; 11:13880. [PMID: 34230555 PMCID: PMC8260734 DOI: 10.1038/s41598-021-93351-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/23/2021] [Indexed: 01/12/2023] Open
Abstract
HER3 is a member of the EGF receptor family and elevated expression is associated with cancer progression and therapy resistance. HER3-specific T-cell engagers might be a suitable treatment option to circumvent the limited efficacy observed for HER3-blocking antibodies in clinical trials. In this study, we developed bispecific antibodies for T-cell retargeting to HER3-expressing tumor cells, utilizing either a single-chain diabody format (scDb) with one binding site for HER3 and one for CD3 on T-cells or a trivalent bispecific scDb-scFv fusion protein exhibiting an additional binding site for HER3. The scDb-scFv showed increased binding to HER3-expressing cancer cell lines compared to the scDb and consequently more effective T-cell activation and T-cell proliferation. Furthermore, the bivalent binding mode of the scDb-scFv for HER3 translated into more potent T-cell mediated cancer cell killing, and allowed to discriminate between moderate and low HER3-expressing target cells. Thus, our study demonstrated the applicability of HER3 for T-cell retargeting with bispecific antibodies, even at moderate expression levels, and the increased potency of an avidity-mediated specificity gain, potentially resulting in a wider safety window of bispecific T-cell engaging antibodies targeting HER3.
Collapse
|
41
|
Ptáková N, Martínek P, Holubec L, Janovský V, Vančurová J, Grossmann P, Navarro PA, Rodriguez Moreno JF, Alaghehbandan R, Hes O, Májek O, Pešek M, Michal M, Ondič O. Identification of tumors with NRG1 rearrangement, including a novel putative pathogenic UNC5D-NRG1 gene fusion in prostate cancer by data-drilling a de-identified tumor database. Genes Chromosomes Cancer 2021; 60:474-481. [PMID: 33583086 DOI: 10.1002/gcc.22942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 02/02/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
The fusion genes containing neuregulin-1 (NRG1) are newly described potentially actionable oncogenic drivers. Initial clinical trials have shown a positive response to targeted treatment in some cases of NRG1 rearranged lung adenocarcinoma, cholangiocarcinoma, and pancreatic carcinoma. The cost-effective large scale identification of NRG1 rearranged tumors is an open question. We have tested a data-drilling approach by performing a retrospective assessment of a de-identified molecular profiling database of 3263 tumors submitted for fusion testing. Gene fusion detection was performed by RNA-based targeted next-generation sequencing using the Archer Fusion Plex kits for Illumina (ArcherDX Inc., Boulder, CO). Novel fusion transcripts were confirmed by a custom-designed RT-PCR. Also, the aberrant expression of CK20 was studied immunohistochemically. The frequency of NRG1 rearranged tumors was 0.2% (7/3263). The most common histologic type was lung adenocarcinoma (n = 5). Also, renal carcinoma (n = 1) and prostatic adenocarcinoma (n = 1) were found. Identified fusion partners were of a wide range (CD74, SDC4, TNC, VAMP2, UNC5D), with CD74, SDC4 being found twice. The UNC5D is a novel fusion partner identified in prostate adenocarcinoma. There was no co-occurrence with the other tested fusions nor KRAS, BRAF, and the other gene mutations specified in the applied gene panels. Immunohistochemically, the focal expression of CK20 was present in 2 lung adenocarcinomas. We believe it should be considered as an incidental finding. In conclusion, the overall frequency of tumors with NRG1 fusion was 0.2%. All tumors were carcinomas. We confirm (invasive mucinous) lung adenocarcinoma as being the most frequent tumor presenting NRG1 fusion. Herein novel putative pathogenic gene fusion UNC5D-NRG1 is described. The potential role of immunohistochemistry in tumor identification should be further addressed.
Collapse
Affiliation(s)
- Nikola Ptáková
- Molecular Genetics Department, Bioptická Laboratoř s.r.o., Pilsen, Czech Republic
- Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petr Martínek
- Molecular Genetics Department, Bioptická Laboratoř s.r.o., Pilsen, Czech Republic
| | - Luboš Holubec
- Department of Clinical Oncology, Na Homolce Hospital, Prague, Czech Republic
- Second Department of Internal Medicine, Medical Faculty in Pilsen, Charles University Prague, Pilsen, Czech Republic
| | - Václav Janovský
- Department of Oncology, Hospital České Budějovice, České Budějovice, Czech Republic
| | - Jana Vančurová
- Department of Oncology, Hospital České Budějovice, České Budějovice, Czech Republic
| | - Petr Grossmann
- Molecular Genetics Department, Bioptická Laboratoř s.r.o., Pilsen, Czech Republic
| | - Paloma Alcaraz Navarro
- Department of Pathology, FiHM-Centro Integral Oncológico Hospital de Madrid Clara Campal, Madrid, Spain
| | - Juan F Rodriguez Moreno
- Department of Pathology, FiHM-Centro Integral Oncológico Hospital de Madrid Clara Campal, Madrid, Spain
| | - Reza Alaghehbandan
- Department of Pathology, University of British Columbia, Royal Columbian Hospital, Vancouver, British Columbia, Canada
| | - Ondřej Hes
- Department of Pathology, Medical Faculty in Pilsen, Charles University Prague, Pilsen, Czech Republic
| | - Ondřej Májek
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Miloš Pešek
- Department of Pneumology and Phthisiology, Medical Faculty in Pilsen, Charles University Prague, Pilsen, Czech Republic
| | - Michal Michal
- Department of Pathology, Medical Faculty in Pilsen, Charles University Prague, Pilsen, Czech Republic
| | - Ondrej Ondič
- Molecular Genetics Department, Bioptická Laboratoř s.r.o., Pilsen, Czech Republic
- Department of Pathology, Medical Faculty in Pilsen, Charles University Prague, Pilsen, Czech Republic
| |
Collapse
|
42
|
Azadi A, Golchini A, Delazar S, Abarghooi Kahaki F, Dehnavi SM, Payandeh Z, Eyvazi S. Recent Advances on Immune Targeted Therapy of Colorectal Cancer Using bi-Specific Antibodies and Therapeutic Vaccines. Biol Proced Online 2021; 23:13. [PMID: 34193050 PMCID: PMC8245152 DOI: 10.1186/s12575-021-00147-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/12/2021] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is a universal heterogeneous disease that is characterized by genetic and epigenetic alterations. Immunotherapy using monoclonal antibodies (mAb) and cancer vaccines are substitute strategies for CRC treatment. When cancer immunotherapy is combined with chemotherapy, surgery, and radiotherapy, the CRC treatment would become excessively efficient. One of the compelling immunotherapy approaches to increase the efficiency of CRC therapy is the deployment of therapeutic mAbs, nanobodies, bi-specific antibodies and cancer vaccines, which improve clinical outcomes in patients. Also, among the possible therapeutic approaches for CRC patients, gene vaccines in combination with antibodies are recently introduced as a new perspective. Here, we aimed to present the current progress in CRC immunotherapy, especially using Bi-specific antibodies and dendritic cells mRNA vaccines. For this aim, all data were extracted from Google Scholar, PubMed, Scopus, and Elsevier, using keywords cancer vaccines; CRC immunotherapy and CRC mRNA vaccines. About 97 articles were selected and investigated completely based on the latest developments and novelties on bi-specific antibodies, mRNA vaccines, nanobodies, and MGD007.
Collapse
Affiliation(s)
- Ali Azadi
- Department of Medicine, De La Salle Health Sciences Institute, Dasmariñas, Philippines
| | - Alireza Golchini
- Cancer surgery Department; Shiraz Medical School, Shiraz University of medical Sciences, Shiraz, Iran
| | - Sina Delazar
- Department of Radiology, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Abarghooi Kahaki
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mohsen Dehnavi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Zahra Payandeh
- Immunology Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Eyvazi
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
- Biotechnology Research Center, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| |
Collapse
|
43
|
Personalized therapeutic strategies in HER2-driven gastric cancer. Gastric Cancer 2021; 24:897-912. [PMID: 33755862 DOI: 10.1007/s10120-021-01165-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Trastuzumab is the only approved targeted therapy in patients with HER2-amplified metastatic gastric cancer (GC). Regrettably, in clinical practice, only a fraction of them achieves long-term benefit from trastuzumab-based upfront strategy. To advance precision oncology, we investigated the therapeutic efficacy of different HER2-targeted strategies, in HER2 "hyper"-amplified (≥ 8 copies) tumors. METHODS We undertook a prospective evaluation of HER2 targeting with monoclonal antibodies, tyrosine kinase inhibitors and antibody-drug conjugates, in a selected subgroup of HER2 "hyper"-amplified gastric patient-derived xenografts (PDXs), through the design of ad hoc preclinical trials. RESULTS Despite the high level of HER2 amplification, trastuzumab elicited a partial response only in 2 out of 8 PDX models. The dual-HER2 blockade with trastuzumab plus either pertuzumab or lapatinib led to complete and durable responses in 5 (62.5%) out of 8 models, including one tumor bearing a concomitant HER2 mutation. In a resistant PDX harboring KRAS amplification, the novel antibody-drug conjugate trastuzumab deruxtecan (but not trastuzumab emtansine) overcame KRAS-mediated resistance. We also identified a HGF-mediated non-cell-autonomous mechanism of secondary resistance to anti-HER2 drugs, responsive to MET co-targeting. CONCLUSION These preclinical randomized trials clearly indicate that in HER2-driven gastric tumors, a boosted HER2 therapeutic blockade is required for optimal efficacy, leading to complete and durable responses in most of the cases. Our results suggest that a selected subpopulation of HER2-"hyper"-amplified GC patients could strongly benefit from this strategy. Despite the negative results of clinical trials, the dual blockade should be reconsidered for patients with clearly HER2-addicted cancers.
Collapse
|
44
|
Radom F, Vonrhein C, Mittl PRE, Plückthun A. Crystal structures of HER3 extracellular domain 4 in complex with the designed ankyrin-repeat protein D5. Acta Crystallogr F Struct Biol Commun 2021; 77:192-201. [PMID: 34196609 PMCID: PMC8248824 DOI: 10.1107/s2053230x21006002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/09/2021] [Indexed: 11/10/2022] Open
Abstract
The members of the human epidermal growth factor receptor (HER) family are among the most intensely studied oncological targets. HER3 (ErbB3), which had long been neglected, has emerged as a key oncogene, regulating the activity of other receptors and being involved in progression and tumor escape in multiple types of cancer. Designed ankyrin-repeat proteins (DARPins) serve as antibody mimetics that have proven to be useful in the clinic, in diagnostics and in research. DARPins have previously been selected against EGFR (HER1), HER2 and HER4. In particular, their combination into bivalent binders that separate or lock receptors in their inactive conformation has proved to be a promising strategy for the design of potent anticancer therapeutics. Here, the selection of DARPins targeting extracellular domain 4 of HER3 (HER3d4) is described. One of the selected DARPins, D5, in complex with HER3d4 crystallized in two closely related crystal forms that diffracted to 2.3 and 2.0 Å resolution, respectively. The DARPin D5 epitope comprises HER3d4 residues 568-577. These residues also contribute to interactions within the tethered (inactive) and extended (active) conformations of the extracellular domain of HER3.
Collapse
Affiliation(s)
- Filip Radom
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Clemens Vonrhein
- Global Phasing Ltd, Sheraton House, Castle Park, Cambridge CB3 0AX, United Kingdom
| | - Peer R. E. Mittl
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| |
Collapse
|
45
|
Corti C, Giugliano F, Nicolò E, Ascione L, Curigliano G. Antibody-Drug Conjugates for the Treatment of Breast Cancer. Cancers (Basel) 2021; 13:2898. [PMID: 34207890 PMCID: PMC8229763 DOI: 10.3390/cancers13122898] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/07/2021] [Accepted: 06/07/2021] [Indexed: 12/15/2022] Open
Abstract
Metastatic breast cancer (BC) is currently an incurable disease. Besides endocrine therapy and targeted agents, chemotherapy is often used in the treatment of this disease. However, lack of tumor specificity and toxicity associated with dose exposure limit the manageability of cytotoxic agents. Antibody-drug conjugates (ADCs) are a relatively new class of anticancer drugs. By merging the selectivity of monoclonal antibodies with the cytotoxic properties of chemotherapy, they improve the therapeutic index of antineoplastic agents. Three core components characterize ADCs: the antibody, directed to a target antigen; the payload, typically a cytotoxic agent; a linker, connecting the antibody to the payload. The most studied target antigen is HER2 with some agents, such as trastuzumab deruxtecan, showing activity not only in HER2-positive, but also in HER2-low BC patients, possibly due to a bystander effect. This property to provide a cytotoxic impact also against off-target cancer cells may overcome the intratumoral heterogeneity of some target antigens. Other cancer-associated antigens represent a strategy for the development of ADCs against triple-negative BC, as shown by the recent approval of sacituzumab govitecan. In this review, we discuss the current landscape of ADC development for the treatment of BC, as well as the possible limitations of this treatment.
Collapse
Affiliation(s)
- Chiara Corti
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (C.C.); (F.G.); (E.N.); (L.A.)
- Department of Oncology and Haematology (DIPO), University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Federica Giugliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (C.C.); (F.G.); (E.N.); (L.A.)
- Department of Oncology and Haematology (DIPO), University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Eleonora Nicolò
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (C.C.); (F.G.); (E.N.); (L.A.)
- Department of Oncology and Haematology (DIPO), University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Liliana Ascione
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (C.C.); (F.G.); (E.N.); (L.A.)
- Department of Oncology and Haematology (DIPO), University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (C.C.); (F.G.); (E.N.); (L.A.)
- Department of Oncology and Haematology (DIPO), University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| |
Collapse
|
46
|
Basu A, Ramamoorthi G, Albert G, Gallen C, Beyer A, Snyder C, Koski G, Disis ML, Czerniecki BJ, Kodumudi K. Differentiation and Regulation of T H Cells: A Balancing Act for Cancer Immunotherapy. Front Immunol 2021; 12:669474. [PMID: 34012451 PMCID: PMC8126720 DOI: 10.3389/fimmu.2021.669474] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/19/2021] [Indexed: 12/22/2022] Open
Abstract
Current success of immunotherapy in cancer has drawn attention to the subsets of TH cells in the tumor which are critical for activation of anti-tumor response either directly by themselves or by stimulating cytotoxic T cell activity. However, presence of immunosuppressive pro-tumorigenic TH subsets in the tumor milieu further contributes to the complexity of regulation of TH cell-mediated immune response. In this review, we present an overview of the multifaceted positive and negative effects of TH cells, with an emphasis on regulation of different TH cell subtypes by various immune cells, and how a delicate balance of contradictory signals can influence overall success of cancer immunotherapy. We focus on the regulatory network that encompasses dendritic cell-induced activation of CD4+ TH1 cells and subsequent priming of CD8+ cytotoxic T cells, along with intersecting anti-inflammatory and pro-tumorigenic TH2 cell activity. We further discuss how other tumor infiltrating immune cells such as immunostimulatory TH9 and Tfh cells, immunosuppressive Treg cells, and the duality of TH17 function contribute to tip the balance of anti- vs pro-tumorigenic TH responses in the tumor. We highlight the developing knowledge of CD4+ TH1 immune response against neoantigens/oncodrivers, impact of current immunotherapy strategies on CD4+ TH1 immunity, and how opposing action of TH cell subtypes can be explored further to amplify immunotherapy success in patients. Understanding the nuances of CD4+ TH cells regulation and the molecular framework undergirding the balancing act between anti- vs pro-tumorigenic TH subtypes is critical for rational designing of immunotherapies that can bypass therapeutic escape to maximize the potential of immunotherapy.
Collapse
Affiliation(s)
- Amrita Basu
- Clinical Science Division, Moffitt Cancer Center, Tampa, FL, United States
| | | | - Gabriella Albert
- Clinical Science Division, Moffitt Cancer Center, Tampa, FL, United States
| | - Corey Gallen
- Clinical Science Division, Moffitt Cancer Center, Tampa, FL, United States
| | - Amber Beyer
- Clinical Science Division, Moffitt Cancer Center, Tampa, FL, United States
| | - Colin Snyder
- Clinical Science Division, Moffitt Cancer Center, Tampa, FL, United States
| | - Gary Koski
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Mary L Disis
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA, United States
| | - Brian J Czerniecki
- Clinical Science Division, Moffitt Cancer Center, Tampa, FL, United States.,Department of Oncological Sciences, University of South Florida, Tampa, FL, United States.,Department of Breast Cancer Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Krithika Kodumudi
- Clinical Science Division, Moffitt Cancer Center, Tampa, FL, United States.,Department of Biological Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
47
|
Capone E, Lattanzio R, Gasparri F, Orsini P, Rossi C, Iacobelli V, De Laurenzi V, Natali PG, Valsasina B, Iacobelli S, Sala G. EV20/NMS-P945, a Novel Thienoindole Based Antibody-Drug Conjugate Targeting HER-3 for Solid Tumors. Pharmaceutics 2021; 13:pharmaceutics13040483. [PMID: 33918158 PMCID: PMC8066800 DOI: 10.3390/pharmaceutics13040483] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
HER-3 is becoming an attractive target for antibody-drug conjugate (ADC)-based therapy. Indeed, this receptor and its ligands are found to be overexpressed in several malignancies, and re-activation of its downstream signaling axis is known to play a critical role in modulating the sensitivity of targeted therapeutics in different tumors. In this study, we generated a novel ADC named EV20/NMS-P945 by coupling the anti-HER-3 antibody EV20 with a duocarmycin-like derivative, the thienoindole (TEI) NMS-P528, a DNA minor groove alkylating agent through a peptidic cleavable linker. This ADC showed target-dependent cytotoxic activity in vitro on several tumor cell lines and therapeutic activity in mouse xenograft tumor models, including those originating from pancreatic, prostatic, head and neck, gastric and ovarian cancer cells and melanoma. Pharmacokinetics and toxicological studies in monkeys demonstrated that this ADC possesses a favorable terminal half-life and stability and it is well tolerated. These data support further EV20/NMS-P945 clinical development as a therapeutic agent against HER-3-expressing malignancies.
Collapse
Affiliation(s)
- Emily Capone
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy; (E.C.); (R.L.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), Via Polacchi 11, 66100 Chieti, Italy;
| | - Rossano Lattanzio
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy; (E.C.); (R.L.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), Via Polacchi 11, 66100 Chieti, Italy;
| | - Fabio Gasparri
- Nerviano Medical Sciences Srl, 20014 Milan, Italy; (F.G.); (P.O.); (B.V.)
| | - Paolo Orsini
- Nerviano Medical Sciences Srl, 20014 Milan, Italy; (F.G.); (P.O.); (B.V.)
| | - Cosmo Rossi
- Center for Advanced Studies and Technology (CAST), Via Polacchi 11, 66100 Chieti, Italy;
| | - Valentina Iacobelli
- Department of Gynecology and Obstetrics, Catholic University, 00168 Rome, Italy;
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy; (E.C.); (R.L.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), Via Polacchi 11, 66100 Chieti, Italy;
| | | | - Barbara Valsasina
- Nerviano Medical Sciences Srl, 20014 Milan, Italy; (F.G.); (P.O.); (B.V.)
| | - Stefano Iacobelli
- MediaPharma s.r.l., Via della Colonnetta 50/A, 66100 Chieti, Italy;
- Correspondence: or (S.I.); (G.S.); Tel.: +39-08-7154-1504 (G.S.)
| | - Gianluca Sala
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy; (E.C.); (R.L.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), Via Polacchi 11, 66100 Chieti, Italy;
- Correspondence: or (S.I.); (G.S.); Tel.: +39-08-7154-1504 (G.S.)
| |
Collapse
|
48
|
Rinne SS, Orlova A, Tolmachev V. PET and SPECT Imaging of the EGFR Family (RTK Class I) in Oncology. Int J Mol Sci 2021; 22:ijms22073663. [PMID: 33915894 PMCID: PMC8036874 DOI: 10.3390/ijms22073663] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
The human epidermal growth factor receptor family (EGFR-family, other designations: HER family, RTK Class I) is strongly linked to oncogenic transformation. Its members are frequently overexpressed in cancer and have become attractive targets for cancer therapy. To ensure effective patient care, potential responders to HER-targeted therapy need to be identified. Radionuclide molecular imaging can be a key asset for the detection of overexpression of EGFR-family members. It meets the need for repeatable whole-body assessment of the molecular disease profile, solving problems of heterogeneity and expression alterations over time. Tracer development is a multifactorial process. The optimal tracer design depends on the application and the particular challenges of the molecular target (target expression in tumors, endogenous expression in healthy tissue, accessibility). We have herein summarized the recent preclinical and clinical data on agents for Positron Emission Tomography (PET) and Single Photon Emission Tomography (SPECT) imaging of EGFR-family receptors in oncology. Antibody-based tracers are still extensively investigated. However, their dominance starts to be challenged by a number of tracers based on different classes of targeting proteins. Among these, engineered scaffold proteins (ESP) and single domain antibodies (sdAb) show highly encouraging results in clinical studies marking a noticeable trend towards the use of smaller sized agents for HER imaging.
Collapse
Affiliation(s)
- Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden; (S.S.R.); (A.O.)
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden; (S.S.R.); (A.O.)
- Science for Life Laboratory, Uppsala University, 752 37 Uppsala, Sweden
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Vladimir Tolmachev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 37 Uppsala, Sweden
- Correspondence: ; Tel.: +46-704-250-782
| |
Collapse
|
49
|
Al-Akhrass H, Conway JRW, Poulsen ASA, Paatero I, Kaivola J, Padzik A, Andersen OM, Ivaska J. A feed-forward loop between SorLA and HER3 determines heregulin response and neratinib resistance. Oncogene 2021; 40:1300-1317. [PMID: 33420373 PMCID: PMC7892347 DOI: 10.1038/s41388-020-01604-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/23/2020] [Accepted: 12/03/2020] [Indexed: 01/29/2023]
Abstract
Current evidence indicates that resistance to the tyrosine kinase-type cell surface receptor (HER2)-targeted therapies is frequently associated with HER3 and active signaling via HER2-HER3 dimers, particularly in the context of breast cancer. Thus, understanding the response to HER2-HER3 signaling and the regulation of the dimer is essential to decipher therapy relapse mechanisms. Here, we investigate a bidirectional relationship between HER2-HER3 signaling and a type-1 transmembrane sorting receptor, sortilin-related receptor (SorLA; SORL1). We demonstrate that heregulin-mediated signaling supports SorLA transcription downstream of the mitogen-activated protein kinase pathway. In addition, we demonstrate that SorLA interacts directly with HER3, forming a trimeric complex with HER2 and HER3 to attenuate lysosomal degradation of the dimer in a Ras-related protein Rab4-dependent manner. In line with a role for SorLA in supporting the stability of the HER2 and HER3 receptors, loss of SorLA compromised heregulin-induced cell proliferation and sensitized metastatic anti-HER2 therapy-resistant breast cancer cells to neratinib in cancer spheroids in vitro and in vivo in a zebrafish brain xenograft model.
Collapse
Affiliation(s)
- Hussein Al-Akhrass
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland.
| | - James R W Conway
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Annemarie Svane Aavild Poulsen
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of biomedicine, Aarhus University, Aarhus, Denmark
| | - Ilkka Paatero
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Jasmin Kaivola
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Artur Padzik
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Olav M Andersen
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of biomedicine, Aarhus University, Aarhus, Denmark
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland.
| |
Collapse
|
50
|
Criscitiello C, Morganti S, Curigliano G. Antibody-drug conjugates in solid tumors: a look into novel targets. J Hematol Oncol 2021; 14:20. [PMID: 33509252 PMCID: PMC7844898 DOI: 10.1186/s13045-021-01035-z] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are a relatively new class of anticancer agents designed to merge the selectivity of monoclonal antibodies with cell killing properties of chemotherapy. They are commonly described as the "Trojan Horses" of therapeutic armamentarium, because of their capability of directly conveying cytotoxic drug (payloads) into the tumor space, thus transforming chemotherapy into a targeted agent. Three novel ADCs have been recently approved, i.e., trastuzumab deruxtecan, sacituzumab govitecan and enfortumab vedotin, respectively, targeting HER2, Trop2 and Nectin4. Thanks to progressive advances in engineering technologies these drugs rely on, the spectrum of diseases sensitive to these drugs as well as their indications are in continuous expansion. Several novel ADCs are under evaluation, exploring new potential targets along with innovative payloads. This review aims at providing a summary of the technology behind these compounds and at presenting the latest ADCs approved in solid tumors, as well as at describing novel targets for ADCs under investigation and new strategies to optimize their efficacy in solid tumors.
Collapse
Affiliation(s)
- Carmen Criscitiello
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Stefania Morganti
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141, Milan, Italy.
- Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy.
| |
Collapse
|