1
|
Shirbhate E, Singh V, Kore R, Koch B, Veerasamy R, Tiwari AK, Rajak H. Synergistic strategies: histone deacetylase inhibitors and platinum-based drugs in cancer therapy. Expert Rev Anticancer Ther 2025:1-21. [PMID: 39873641 DOI: 10.1080/14737140.2025.2458156] [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: 09/20/2024] [Revised: 12/23/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
INTRODUCTION The synergistic combination of histone deacetylase inhibitors and platinum-based medicines represents a promising therapeutic strategy to efficacy and overcome drug resistance in cancer therapy, necessitating a comprehensive understanding on their molecular interactions and clinical potential. AREAS COVERED The objective of presented review is to investigate the molecular pathways of platinum medicines and HDAC inhibitors. A comprehensive literature review from 2011 to 2024 was conducted across multiple databases like MEDLINE, PubMed, Google Scholar, Science Direct, Scopus and official websites of ClinicalTrial.gov to explore publications on HDAC inhibitors, platinum drugs, and combination cancer therapies, revealing preliminary evidence of innovative treatment strategies involving HDAC inhibitors and platinum chemotherapeutics. Several new platinum (IV) complexes, with HDAC inhibitory moieties and better cytotoxicity profiles than conventional platinum drugs, are also reviewed here. EXPERT OPINION The above combination has great potential in cancer treatment, however managing toxicity, dosage regimens, and patient selection biomarkers are problematic. More selective HDAC inhibitors and innovative delivery techniques are potential areas for future research. An adaptation toward changing cancer therapeutic landscapes, highlights combining HDAC inhibitors with platinum-based medicines serves as a new concept for personalized medicine, however, a deeper research is still needed at this time.
Collapse
Affiliation(s)
- Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, India
| | - Vaibhav Singh
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, India
| | - Rakesh Kore
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, India
| | - Biplab Koch
- Department of Zoology, Banaras Hindu University, Varanasi, India
| | | | - Amit Kumar Tiwari
- Cancer & System Therapeutics, UAMS College of Pharmacy, UAMS - University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, India
| |
Collapse
|
2
|
Abed R, Alhroub W, Asbeh YA, Rabee A, Bannoura S, Madia A. A rare presentation of primary lung adenocarcinoma mimicking bilateral interstitial infiltration: A case report and literature review. Int J Surg Case Rep 2025; 127:110899. [PMID: 39823974 PMCID: PMC11786693 DOI: 10.1016/j.ijscr.2025.110899] [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: 12/13/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Primary lung adenocarcinoma can sometimes present atypically, mimicking interstitial lung disease (ILD), and posing significant diagnostic challenges. Such presentations often lead to misdiagnoses, delaying appropriate treatment. CASE PRESENTATION A 35-year-old female non-smoker presented with a six-month history of progressive cough, mild hemoptysis, fatigue, and exertional dyspnea, with no associated weight loss. Imaging studies revealed diffuse ground-glass opacities and interstitial infiltrates, while pulmonary function tests were consistent with interstitial lung disease. Despite these findings, bronchoscopy results were normal. A definitive diagnosis was ultimately made through a biopsy, which identified a moderately to poorly differentiated adenocarcinoma with acinar and micropapillary features. DISCUSSION This case highlights the diagnostic complexity when lung adenocarcinoma presents atypically, mimicking ILD. Conventional diagnostic tools, such as imaging and pulmonary function tests, may overlap with ILD findings, leading to misdiagnoses. Early consideration of malignancy and the use of invasive diagnostic procedures, such as biopsy, are essential for distinguishing between ILD and malignancy in atypical cases. CONCLUSION This case underscores the importance of maintaining a high index of suspicion for malignancy in atypical ILD presentations. Early invasive diagnostic techniques are crucial for achieving a timely and accurate diagnosis, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Renad Abed
- Faculty of Medicine, Hebron University, Hebron, Palestine
| | - Wasef Alhroub
- Faculty of Medicine, Hebron University, Hebron, Palestine.
| | | | | | - Sami Bannoura
- Department of pathology, Al-Ahli hospital, Hebron, Palestine
| | | |
Collapse
|
3
|
Okuno T, Isobe T, Tsubata Y. Current pharmacologic treatment of brain metastasis in non-small cell lung cancer. Clin Exp Metastasis 2024; 41:549-565. [PMID: 38466521 PMCID: PMC11499348 DOI: 10.1007/s10585-024-10276-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/28/2024] [Indexed: 03/13/2024]
Abstract
Lung cancer is a type of cancer that can metastasize to the lungs, brain, bones, liver, adrenal glands, and other organs; however, the occurrence of brain metastases is the most common event. Symptoms of brain metastasis include motor dysfunction, mental dysfunction, seizures, headaches, nausea, and vomiting, and significantly reduce the quality of life of cancer patients. Brain metastases are a poor prognostic factor, and controlling them is extremely important for prolonging prognosis and improving the quality of life. Currently, local surgery and radiotherapy are recommended for their treatment. However, recently, cancer treatments using molecular-targeted drugs and immune checkpoint inhibitors have been introduced, which may also be effective against brain metastases. Therefore, it is necessary to determine whether local or systemic therapy is optimal for each case. In this review, we focus on recent findings regarding drug therapy in treating brain metastases from advanced non-small cell lung cancer.
Collapse
Affiliation(s)
- Takae Okuno
- Division of Medical Oncology and Respiratory Medicine, Department of Internal Medicine, Shimane University Faculty of Medicine, 89-1, Enyacho, Izumo, Shimane, 693-8501, Japan
| | - Takeshi Isobe
- Division of Medical Oncology and Respiratory Medicine, Department of Internal Medicine, Shimane University Faculty of Medicine, 89-1, Enyacho, Izumo, Shimane, 693-8501, Japan
| | - Yukari Tsubata
- Division of Medical Oncology and Respiratory Medicine, Department of Internal Medicine, Shimane University Faculty of Medicine, 89-1, Enyacho, Izumo, Shimane, 693-8501, Japan.
| |
Collapse
|
4
|
Brunetti B, de Biase D, Millanta F, Muscatello LV, Di Oto E, Marchetti R, Laddaga EL, De Leo A, Tallini G, Bacci B. Protein Expression, Amplification, and Mutation of HER2 Gene in Canine Primary Pulmonary Adenocarcinomas: Preliminary Results. Animals (Basel) 2024; 14:2625. [PMID: 39335216 PMCID: PMC11428963 DOI: 10.3390/ani14182625] [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: 07/04/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Recently, human epidermal growth factor receptor 2 (HER2) has emerged as a therapeutic target of interest for non-small-cell lung cancer in humans. The role of HER2 in canine pulmonary adenocarcinomas is poorly documented. To address this gap, this study employed three methodologies: immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), and next-generation sequencing (NGS) to investigate the protein expression, gene amplification, and mutation of HER2 in 19 canine primary pulmonary adenocarcinomas. By IHC, 3 out of 19 cases were overexpressed 3+, 6 were 2+, and 10 were negative. With FISH, 2 cases were amplified (12.5%), 3 were inadequate for the analyses, and the others were non-amplified. With NGS, seven cases were inadequate. All other cases were wild-type, except for one IHC 3+ case, which was amplified with FISH and with a specific mutation already described in human pulmonary adenocarcinoma, V659E. This mutation is probably sensitive to tyrosine kinase inhibitory drugs. These results are similar to those in human medicine and to the few data in the literature on canine lung carcinomas; the presence of 12.5% of amplified cases in dogs lays the foundation for future targeted drugs against HER2 alterations.
Collapse
Affiliation(s)
- Barbara Brunetti
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Dario de Biase
- Department of Experimental, Diagnostics and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
| | - Francesca Millanta
- Department of Veterinary Sciences, University of Pisa, 56126 Pisa, Italy
| | - Luisa Vera Muscatello
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy
| | | | - Roberta Marchetti
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Ester Lidia Laddaga
- Ospedale Veterinario "I Portoni Rossi", Anicura, Zola Predosa, 40069 Bologna, Italy
| | - Antonio De Leo
- Department of Experimental, Diagnostics and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40126 Bologna, Italy
| | - Giovanni Tallini
- Department of Experimental, Diagnostics and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40126 Bologna, Italy
| | - Barbara Bacci
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
5
|
Pan X, Zhou X. Long term survival achieved through combination of almonertinib and pyrotinib in EGFR-mutant/HER2-amplified advanced NSCLC patient: a case report and literature review. Front Oncol 2024; 14:1397238. [PMID: 39184039 PMCID: PMC11341367 DOI: 10.3389/fonc.2024.1397238] [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: 03/07/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
Backgroud Human epithelial growth factor receptor 2 (HER2) amplification is an important mechanism of acquired resistance to anti-epidermal growth factor receptor (EGFR) therapy in non-small cell lung cancer (NSCLC) patients. For patients with both EGFR mutation and HER2 amplification, there is currently no unified standard treatment, and further exploration is needed on how to choose the therapy. Methods and results A female NSCLC patient developed bone and brain metastases 14 and 42 months after radical surgery, respectively. The second genetic sequencing detected EGFR L858R mutation and HER2 amplification, and therefore initiated treatment with almonertinib and pyrotinib. The patient achieved partial remission and did not show any further progression during the follow-up period. Conclusion For NSCLC patients with both EGFR mutation and HER2 amplification, the combination of almonertinib and pyrotinib is a valuable therapy that can continuously reduce tumor burden and achieve long-term survival.
Collapse
Affiliation(s)
| | - Xiao Zhou
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
6
|
Wu Y, Yu G, Jin K, Qian J. Advancing non-small cell lung cancer treatment: the power of combination immunotherapies. Front Immunol 2024; 15:1349502. [PMID: 39015563 PMCID: PMC11250065 DOI: 10.3389/fimmu.2024.1349502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) remains an unsolved challenge in oncology, signifying a substantial global health burden. While considerable progress has been made in recent years through the emergence of immunotherapy modalities, such as immune checkpoint inhibitors (ICIs), monotherapies often yield limited clinical outcomes. The rationale behind combining various immunotherapeutic or other anticancer agents, the mechanistic underpinnings, and the clinical evidence supporting their utilization is crucial in NSCLC therapy. Regarding the synergistic potential of combination immunotherapies, this study aims to provide insights to help the landscape of NSCLC treatment and improve clinical outcomes. In addition, this review article discusses the challenges and considerations of combination regimens, including toxicity management and patient selection.
Collapse
Affiliation(s)
- Yuanlin Wu
- Department of Thoracic Surgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Guangmao Yu
- Department of Thoracic Surgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Ketao Jin
- Department of Gastrointestinal, Colorectal and Anal Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People’s Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang, Zhejiang, China
| |
Collapse
|
7
|
Zhang H, Finkelman BS, Ettel MG, Velez MJ, Turner BM, Hicks DG. HER2 evaluation for clinical decision making in human solid tumours: pearls and pitfalls. Histopathology 2024; 85:3-19. [PMID: 38443321 DOI: 10.1111/his.15170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/23/2024] [Accepted: 02/17/2024] [Indexed: 03/07/2024]
Abstract
The significant clinical benefits of human epidermal growth factor receptor 2 (HER2)-targeted therapeutic agents have revolutionized the clinical treatment landscape in a variety of human solid tumours. Accordingly, accurate evaluation of HER2 status in these different tumour types is critical for clinical decision making to select appropriate patients who may benefit from life-saving HER2-targeted therapies. HER2 biomarker scoring criteria is different in different organ systems, and close adherence to the corresponding HER2 biomarker testing guidelines and their updates, if available, is essential for accurate evaluation. In addition, knowing the unusual patterns of HER2 expression is also important to avoid inaccurate evaluation. In this review, we discuss the key considerations when evaluating HER2 status in solid tumours for clinical decision making, including tissue handling and preparation for HER2 biomarker testing, as well as pathologist's readout of HER2 testing results in breast carcinomas, gastroesophageal adenocarcinomas, colorectal adenocarcinomas, gynaecologic carcinomas, and non-small cell lung carcinomas.
Collapse
Affiliation(s)
- Huina Zhang
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Brian S Finkelman
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Mark G Ettel
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Moises J Velez
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Bradley M Turner
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - David G Hicks
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
8
|
Lee Y, Lee B, Choi YL, Kang DW, Han J. Clinicopathologic and Molecular Characteristics of HER2 (ERBB2)-Altered Non-Small Cell Lung Cancer: Implications for Precision Medicine. Mod Pathol 2024; 37:100490. [PMID: 38588887 DOI: 10.1016/j.modpat.2024.100490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 03/18/2024] [Accepted: 03/30/2024] [Indexed: 04/10/2024]
Abstract
The heterogeneous relationship between protein expression, amplification, and mutations in human epidermal growth factor receptor 2 (HER2) in non-small cell lung cancer (NSCLC) and the optimal methods for detecting these alterations remain unclear. We aimed to elucidate the clinicopathological and molecular characteristics of HER2-altered NSCLC and investigate practical approaches for identifying patients who might benefit from HER2-targeted therapies. Using next-generation sequencing data from 1680 individuals, we searched for patients with HER2-altered NSCLCs, including amplifications and mutations. Clinicopathological data and tissue slides were reviewed. Immunohistochemistry (IHC) and silver in situ hybridization were performed according to the American Society of Clinical Oncology/College of American Pathologists guidelines. Our analysis identified 89 (5.3%) patients with HER2-altered NSCLCs, comprising 30 (1.8%) with amplification and 59 (3.6%) mutations, and they were compared with 165 control patients. Of the 59 HER2-mutated cases, 52 harbored tyrosine kinase domain (TKD) mutations, primarily HER2 exon 20 insertions. HER2 TKD alterations were associated with younger age, female sex, nonsmoking status, adenocarcinoma with a micropapillary pattern, lung-to-lung metastasis, and poor overall survival. The 33 patients with TKD mutations and 3 with non-TKD point mutations showed incomplete or complete membranous HER2 immunoreactivity (1+ and 2+, 61.07%). Six patients exhibiting amplifications had an IHC score of ≤2+ despite their high copy numbers and concomitantly displayed other actionable EGFR, KRAS, SMARCA4, and other HER2 mutations. These HER2-altered NSCLCs with molecular coalterations showed heterogeneous patterns through HER2 IHC and silver in situ hybridization. Therefore, next-generation sequencing should be used to identify HER2 mutations in patients with NSCLC who present with concomitant alterations. In addition, the above clinicopathological characteristics and HER2 IHC results can be valuable determinants for identifying patients with HER2-altered NSCLC. These insights hold promise for the development of more effective diagnostic and therapeutic strategies for this complex subset of NSCLC patients.
Collapse
Affiliation(s)
- Yurimi Lee
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Department of Pathology, Chungnam National University College of Medicine, Daejeon, South Korea
| | - Boram Lee
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.
| | - Dong-Wook Kang
- Department of Pathology, Chungnam National University College of Medicine, Daejeon, South Korea
| | - Joungho Han
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
9
|
Kaji K, Motegi T, Yonezawa T, Momoi Y, Maeda S. Anti-tumour effects of lapatinib on HER2-positive canine prostatic carcinoma cell lines. Open Vet J 2024; 14:1259-1268. [PMID: 38938437 PMCID: PMC11199751 DOI: 10.5455/ovj.2024.v14.i5.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/22/2024] [Indexed: 06/29/2024] Open
Abstract
Background Canine prostatic carcinoma (cPC) is a urogenital tumour with a poor prognosis, for which no effective treatment has been established. Recently, it has been shown that human epidermal growth factor receptor type 2 (HER2) is overexpressed in cPC cells; however, the efficacy of HER2-targeted therapy remains unclear. Aim Investigate the anti-tumour effect of lapatinib on HER2-positive cPC cell lines. Methods Two cell lines (muPC and bePC) were established from two dogs with cPC and the effects of lapatinib treatment on cell proliferation, apoptosis, and HER2 downstream signalling were investigated. Furthermore, muPC was used to generate tumour-bearing mice, and the anti-tumour effects of lapatinib were examined in vivo. Results Lapatinib treatment inhibited the proliferation and phosphorylation of Erk1/2 and Akt, which are downstream signals of HER2. Furthermore, the TUNEL assay showed that lapatinib induced apoptosis in both cell lines. The muPC-engrafted nude mouse model showed that lapatinib significantly inhibited tumour growth and increased the area of necrotic tumour tissue compared to the vehicle-treated groups. Conclusion Lapatinib exerts anti-tumour effects on cPC cells by inhibiting HER-2 signalling.
Collapse
Affiliation(s)
- Kenjiro Kaji
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomoki Motegi
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomohiro Yonezawa
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Momoi
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shingo Maeda
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
10
|
Syed RU, Afsar S, Aboshouk NAM, Salem Alanzi S, Abdalla RAH, Khalifa AAS, Enrera JA, Elafandy NM, Abdalla RAH, Ali OHH, Satheesh Kumar G, Alshammari MD. LncRNAs in necroptosis: Deciphering their role in cancer pathogenesis and therapy. Pathol Res Pract 2024; 256:155252. [PMID: 38479121 DOI: 10.1016/j.prp.2024.155252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 04/14/2024]
Abstract
Necroptosis, a controlled type of cell death that is different from apoptosis, has become a key figure in the aetiology of cancer and offers a possible target for treatment. A growing number of biological activities, including necroptosis, have been linked to long noncoding RNAs (lncRNAs), a varied family of RNA molecules with limited capacity to code for proteins. The complex interactions between LncRNAs and important molecular effectors of necroptosis, including mixed lineage kinase domain-like pseudokinase (MLKL) and receptor-interacting protein kinase 3 (RIPK3), will be investigated. We will explore the many methods that LncRNAs use to affect necroptosis, including protein-protein interactions, transcriptional control, and post-transcriptional modification. Additionally, the deregulation of certain LncRNAs in different forms of cancer will be discussed, highlighting their dual function in influencing necroptotic processes as tumour suppressors and oncogenes. The goal of this study is to thoroughly examine the complex role that LncRNAs play in controlling necroptotic pathways and how that regulation affects the onset and spread of cancer. In the necroptosis for cancer treatment, this review will also provide insight into the possible therapeutic uses of targeting LncRNAs. Techniques utilising LncRNA-based medicines show promise in controlling necroptotic pathways to prevent cancer from spreading and improve the effectiveness of treatment.
Collapse
Affiliation(s)
- Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Hail 81442, Saudi Arabia.
| | - S Afsar
- Department of Virology, Sri Venkateswara University, Tirupathi, Andhra Pradesh 517502, India.
| | - Nayla Ahmed Mohammed Aboshouk
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | | | | | - Amna Abakar Suleiman Khalifa
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Jerlyn Apatan Enrera
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Nancy Mohammad Elafandy
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Randa Abdeen Husien Abdalla
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Omar Hafiz Haj Ali
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - G Satheesh Kumar
- Department of Pharmaceutical Chemistry, College of Pharmacy, Seven Hills College of Pharmacy, Venkataramapuram, Tirupati, India
| | - Maali D Alshammari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| |
Collapse
|
11
|
Asencio-Durán M, Fernández-Gutiérrez E, Larrañaga-Cores M, Klein-Burgos C, Dabad-Moreno JV, Capote-Díez M. Ocular side effects of oncological therapies: Review. ARCHIVOS DE LA SOCIEDAD ESPANOLA DE OFTALMOLOGIA 2024; 99:109-132. [PMID: 37949110 DOI: 10.1016/j.oftale.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/26/2023] [Indexed: 11/12/2023]
Abstract
With the advance of cancer therapy in recent years, the knowledge of the mechanisms involved in this disease has increased, which has meant an increase in the quality of life and survival of patients with tumor pathologies previously considered incurable or refractory to treatment. The number of drugs used has increased exponentially in number, and although the implicit toxicity is lower than that of conventional antineoplastic therapy, they lead to the appearance of new associated adverse effects that the ophthalmologist must recognize and manage.
Collapse
Affiliation(s)
- M Asencio-Durán
- Servicio de Oftalmología, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital La Paz (IDIPAZ), Madrid, Spain.
| | - E Fernández-Gutiérrez
- Servicio de Oftalmología, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital La Paz (IDIPAZ), Madrid, Spain
| | - M Larrañaga-Cores
- Servicio de Oftalmología, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital La Paz (IDIPAZ), Madrid, Spain
| | - C Klein-Burgos
- Servicio de Oftalmología, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital La Paz (IDIPAZ), Madrid, Spain
| | - J V Dabad-Moreno
- Servicio de Oftalmología, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital La Paz (IDIPAZ), Madrid, Spain
| | - M Capote-Díez
- Servicio de Oftalmología, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital La Paz (IDIPAZ), Madrid, Spain
| |
Collapse
|
12
|
La’ah AS, Chiou SH. Cutting-Edge Therapies for Lung Cancer. Cells 2024; 13:436. [PMID: 38474400 PMCID: PMC10930724 DOI: 10.3390/cells13050436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Lung cancer remains a formidable global health challenge that necessitates inventive strategies to improve its therapeutic outcomes. The conventional treatments, including surgery, chemotherapy, and radiation, have demonstrated limitations in achieving sustained responses. Therefore, exploring novel approaches encompasses a range of interventions that show promise in enhancing the outcomes for patients with advanced or refractory cases of lung cancer. These groundbreaking interventions can potentially overcome cancer resistance and offer personalized solutions. Despite the rapid evolution of emerging lung cancer therapies, persistent challenges such as resistance, toxicity, and patient selection underscore the need for continued development. Consequently, the landscape of lung cancer therapy is transforming with the introduction of precision medicine, immunotherapy, and innovative therapeutic modalities. Additionally, a multifaceted approach involving combination therapies integrating targeted agents, immunotherapies, or traditional cytotoxic treatments addresses the heterogeneity of lung cancer while minimizing its adverse effects. This review provides a brief overview of the latest emerging therapies that are reshaping the landscape of lung cancer treatment. As these novel treatments progress through clinical trials are integrated into standard care, the potential for more effective, targeted, and personalized lung cancer therapies comes into focus, instilling renewed hope for patients facing challenging diagnoses.
Collapse
Affiliation(s)
- Anita Silas La’ah
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan;
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan;
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
13
|
Popović L, Wintgens JP, Wu Y, Brankatschk B, Menninger S, Degenhart C, Jensen N, Wichert SP, Klebl B, Rossner MJ, Wehr MC. Profiling of ERBB receptors and downstream pathways reveals selectivity and hidden properties of ERBB4 antagonists. iScience 2024; 27:108839. [PMID: 38303712 PMCID: PMC10831936 DOI: 10.1016/j.isci.2024.108839] [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: 10/11/2023] [Revised: 11/20/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
ERBB receptor tyrosine kinases are involved in development and diseases like cancer, cardiovascular, neurodevelopmental, and mental disorders. Although existing drugs target ERBB receptors, the next generation of drugs requires enhanced selectivity and understanding of physiological pathway responses to improve efficiency and reduce side effects. To address this, we developed a multilevel barcoded reporter profiling assay, termed 'ERBBprofiler', in living cells to monitor the activity of all ERBB targets and key physiological pathways simultaneously. This assay helps differentiate on-target therapeutic effects from off-target and off-pathway side effects of ERBB antagonists. To challenge the assay, eight established ERBB antagonists were profiled. Known effects were confirmed, and previously uncharacterized properties were discovered, such as pyrotinib's preference for ERBB4 over EGFR. Additionally, two lead compounds selectively targeting ERBB4 were profiled, showing promise for clinical trials. Taken together, this multiparametric profiling approach can guide early-stage drug development and lead to improved future therapeutic interventions.
Collapse
Affiliation(s)
- Lukša Popović
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| | - Jan P. Wintgens
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| | - Yuxin Wu
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Ben Brankatschk
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| | - Sascha Menninger
- Lead Discovery Center GmbH, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | - Carsten Degenhart
- Lead Discovery Center GmbH, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | - Niels Jensen
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Sven P. Wichert
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Bert Klebl
- Lead Discovery Center GmbH, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | - Moritz J. Rossner
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
| | - Michael C. Wehr
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstrasse 7, 80336 Munich, Germany
- Systasy Bioscience GmbH, Balanstrasse 6, 81669 Munich, Germany
| |
Collapse
|
14
|
Zhang J, Han W, Guo J, Zhang C, Cao L, Peng L, Han X, Wang Z. Efficacy of immunotherapy in HER2-mutated non-small cell lung cancer: a single-arm meta-analysis. J Cancer Res Clin Oncol 2024; 150:42. [PMID: 38280966 PMCID: PMC10822003 DOI: 10.1007/s00432-023-05509-0] [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: 08/02/2023] [Accepted: 11/13/2023] [Indexed: 01/29/2024]
Abstract
BACKGROUND Non-small cell lung cancers (NSCLC) harboring Human Epidermal Growth Factor Receptor 2 (HER2) mutations represent a distinct subset with unique therapeutic challenges. Although immune checkpoint inhibitors (ICIs) have been transformative in lung cancer treatment, the efficacy of ICIs in HER2-mutated NSCLC remains to be established. METHODS We systematically searched for real-world studies investigating the use of ICIs in treating HER2-mutated NSCLC, sourced from the PubMed, Cochrane Library, and Embase databases. Outcomes including objective response rate (ORR), disease control rate (DCR), and progression-free survival (PFS) were extracted for further analysis. RESULTS Twelve studies involving 260 patients were enrolled in this meta-analysis. Pooled data revealed an ORR of 0.26 (95% CI 0.17-0.34), a DCR of 0.68 (95% CI 0.55-0.81), and a median PFS (mPFS) of 5.36 months (95% CI 3.50-7.21). Notably, in the subgroup receiving combined immune and chemotherapy, the ORR increased to 0.37 (95% CI 0.26-0.49), the DCR to 0.79 (95% CI 0.70-0.87), and the mPFS to 7.10 months (95% CI 5.21-8.99). CONCLUSIONS ICIs demonstrate promising anti-tumor activity and safety in patients with HER2-mutated NSCLC. Furthermore, the combined regimen of ICIs and chemotherapy may provide a significant therapeutic option for this patient population.
Collapse
Affiliation(s)
- Juguang Zhang
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Weizhong Han
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jun Guo
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Chufeng Zhang
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lijun Cao
- Department of Oncology, Liaocheng Chiping District Hospital of Traditional Chinese Medicine, Liaocheng, Shandong, China
| | - Lixiu Peng
- Department of Internal Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiao Han
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Zhehai Wang
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
15
|
Shen L, Li Y, Huang H, Lu Z, Chen B. HER2 in Gastric Cancer: A Comprehensive Analysis Combining Meta-Analysis and DCE-MRI Radiomics. Cancer Control 2024; 31:10732748241293699. [PMID: 39448273 PMCID: PMC11528791 DOI: 10.1177/10732748241293699] [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: 12/14/2023] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024] Open
Abstract
OBJECTIVE Advanced gastric cancer (AGC) is a severe malignant tumor, and overexpression of HER2/ERBB2 may play a crucial role in its development. The purpose of this study is to investigate the overexpression of HER2/ERBB2 in gastric cancer through a meta-analysis and examine its relationship with perfusion parameters using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) technology. METHODS We conducted an extensive literature search and collected relevant studies for the meta-analysis. We used a random-effects model and assigned weights using the "inverse variance" method. Additionally, we included 95 AGC patients diagnosed pathologically between April 2018 and October 2021. They all underwent DCE-MRI scans, and the data were subsequently analyzed using the Omni kinetic software. HER2 expression was assessed using immunohistochemistry. RESULTS The meta-analysis revealed an overall odds ratio (OR) of .21 for HER2/ERBB2 overexpression in gastric cancer, with a 95% confidence interval of [.14, .30]. DCE-MRI results showed a significant association between high HER2 expression and poor tumor differentiation (P < .005). The extracellular volume fraction (Ecv) quantile, mean, relative deviation, median intensity, and difference entropy were significantly higher in the low HER2 expression group compared to the high HER2 expression group. Receiver operating characteristic (ROC) curve analysis demonstrated that the area under the curve (AUC) values of DCE-MRI radiomic parameters with significant differences were close to .7. CONCLUSION Overexpression of HER2/ERBB2 in gastric cancer is significantly associated with certain radiomic parameters of DCE-MRI, providing a valuable diagnostic tool for clinical practice. Furthermore, the meta-analysis further confirmed the critical role of HER2/ERBB2 in the development of gastric cancer.
Collapse
Affiliation(s)
- Liyijing Shen
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, P. R. China
| | - Yaoqing Li
- Department of Gastrointestinal Surgery, Shaoxing People’s Hospital, Shaoxing, P. R. China
| | - Huizhen Huang
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, P. R. China
| | - Zengxin Lu
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, P. R. China
| | - Bo Chen
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, P. R. China
| |
Collapse
|
16
|
Žagar Ž, Schmidt JM. A Scoping Review on Tyrosine Kinase Inhibitors in Cats: Current Evidence and Future Directions. Animals (Basel) 2023; 13:3059. [PMID: 37835664 PMCID: PMC10572079 DOI: 10.3390/ani13193059] [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: 08/23/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have become invaluable in the treatment of human and canine malignancies, but their role in feline oncology is less defined. While toceranib phosphate and masitinib mesylate are licensed for use in dogs, no TKI is yet approved for cats. This review systematically maps the research conducted on the expression of tyrosine kinases in neoplastic and non-neoplastic domestic feline tissues, as well as the in vitro/in vivo use of TKIs in domestic cats. We identify and discuss knowledge gaps and speculate on the further research and potential indications for TKI use in cats. A comprehensive search of three electronic databases and relevant paper reference lists identified 139 studies meeting the inclusion criteria. The most commonly identified tumors were mast cell tumors (MCTs), mammary and squamous cell carcinomas and injection-site sarcomas. Based on the current literature, toceranib phosphate appears to be the most efficacious TKI in cats, especially against MCTs. Exploring the clinical use of TKIs in mammary carcinomas holds promise. Despite the progress, currently, the evidence falls short, underscoring the need for further research to discover new indications in feline oncology and to bridge the knowledge gaps between human and feline medicine.
Collapse
Affiliation(s)
- Žiga Žagar
- IVC Evidensia Small Animal Clinic Hofheim, 65719 Hofheim am Taunus, Germany
| | | |
Collapse
|
17
|
Ngaha TYS, Zhilenkova AV, Essogmo FE, Uchendu IK, Abah MO, Fossa LT, Sangadzhieva ZD, D. Sanikovich V, S. Rusanov A, N. Pirogova Y, Boroda A, Rozhkov A, Kemfang Ngowa JD, N. Bagmet L, I. Sekacheva M. Angiogenesis in Lung Cancer: Understanding the Roles of Growth Factors. Cancers (Basel) 2023; 15:4648. [PMID: 37760616 PMCID: PMC10526378 DOI: 10.3390/cancers15184648] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Research has shown the role of growth factors in lung cancer angiogenesis. Angiogenesis promotes lung cancer progression by stimulating tumor growth, enhancing tumor invasion, contributing to metastasis, and modifying immune system responses within the tumor microenvironment. As a result, new treatment techniques based on the anti-angiogenic characteristics of compounds have been developed. These compounds selectively block the growth factors themselves, their receptors, or the downstream signaling pathways activated by these growth factors. The EGF and VEGF families are the primary targets in this approach, and several studies are being conducted to propose anti-angiogenic drugs that are increasingly suitable for the treatment of lung cancer, either as monotherapy or as combined therapy. The efficacy of the results are encouraging, but caution must be placed on the higher risk of toxicity, outlining the importance of personalized follow-up in the management of these patients.
Collapse
Affiliation(s)
- Tchawe Yvan Sinclair Ngaha
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
- Department of Public Health, James Lind Institute, Rue de la Cité 1, 1204 Geneva, Switzerland
| | - Angelina V. Zhilenkova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Freddy Elad Essogmo
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Ikenna K. Uchendu
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
- Medical Laboratory Science Department, Faculty of Health Science and Technology, College of Medicine, University of Nigeria, Enugu Campus, Enugu 410001, Nigeria
| | - Moses Owoicho Abah
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Lionel Tabola Fossa
- Department of Oncology, Bafoussam Regional Hospital, Bafoussam 980, Cameroon;
| | - Zaiana D. Sangadzhieva
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Varvara D. Sanikovich
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Alexander S. Rusanov
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Yuliya N. Pirogova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Alexander Boroda
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Alexander Rozhkov
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Jean D. Kemfang Ngowa
- Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Yaounde 1364, Cameroon;
| | - Leonid N. Bagmet
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Marina I. Sekacheva
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| |
Collapse
|
18
|
Uram Ł, Wróbel K, Walczak M, Szymaszek Ż, Twardowska M, Wołowiec S. Exploring the Potential of Lapatinib, Fulvestrant, and Paclitaxel Conjugated with Glycidylated PAMAM G4 Dendrimers for Cancer and Parasite Treatment. Molecules 2023; 28:6334. [PMID: 37687164 PMCID: PMC10489794 DOI: 10.3390/molecules28176334] [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: 07/16/2023] [Revised: 08/06/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Fulvestrant (F), lapatinib (L), and paclitaxel (P) are hydrophobic, anticancer drugs used in the treatment of estrogen receptor (ER) and epidermal growth factor receptor (EGFR)-positive breast cancer. In this study, glycidylated PAMAM G4 dendrimers, substituted with F, L, and/or P and targeting tumor cells, were synthesized and characterized, and their antitumor activity against glioma U-118 MG and non-small cell lung cancer A549 cells was tested comparatively with human non-tumorogenic keratinocytes (HaCaT). All cell lines were ER+ and EGFR+. In addition, the described drugs were tested in the context of antinematode therapy on C. elegans. The results show that the water-soluble conjugates of G4P, G4F, G4L, and G4PFL actively entered the tested cells via endocytosis due to the positive zeta potential (between 13.57-40.29 mV) and the nanoparticle diameter of 99-138 nm. The conjugates of G4P and G4PFL at nanomolar concentrations were the most active, and the least active conjugate was G4F. The tested conjugates inhibited the proliferation of HaCaT and A549 cells; in glioma cells, cytotoxicity was associated mainly with cell damage (mitochondria and membrane transport). The toxicity of the conjugates was proportional to the number of drug residues attached, with the exception of G4L; its action was two- and eight-fold stronger against glioma and keratinocytes, respectively, than the equivalent of lapatinib alone. Unfortunately, non-cancer HaCaT cells were the most sensitive to the tested constructs, which forced a change in the approach to the use of ER and EGFR receptors as a goal in cancer therapy. In vivo studies on C. elegans have shown that all compounds, most notably G4PFL, may be potentially useful in anthelmintic therapy.
Collapse
Affiliation(s)
- Łukasz Uram
- Faculty of Chemistry, Rzeszów University of Technology, 6 Powstańcow Warszawy Ave., 35-959 Rzeszów, Poland; (Ł.U.); (M.W.); (Ż.S.); (M.T.)
| | - Konrad Wróbel
- Medical College, Rzeszów University, 1a Warzywna Street, 35-310 Rzeszów, Poland;
| | - Małgorzata Walczak
- Faculty of Chemistry, Rzeszów University of Technology, 6 Powstańcow Warszawy Ave., 35-959 Rzeszów, Poland; (Ł.U.); (M.W.); (Ż.S.); (M.T.)
| | - Żaneta Szymaszek
- Faculty of Chemistry, Rzeszów University of Technology, 6 Powstańcow Warszawy Ave., 35-959 Rzeszów, Poland; (Ł.U.); (M.W.); (Ż.S.); (M.T.)
| | - Magdalena Twardowska
- Faculty of Chemistry, Rzeszów University of Technology, 6 Powstańcow Warszawy Ave., 35-959 Rzeszów, Poland; (Ł.U.); (M.W.); (Ż.S.); (M.T.)
| | - Stanisław Wołowiec
- Medical College, Rzeszów University, 1a Warzywna Street, 35-310 Rzeszów, Poland;
| |
Collapse
|
19
|
Chen Y, Zhang S, Lu J, Li D, Wu H, Zhang L, Li X, Gao X, Xu Y, Zeng Z, Zeng L, Ding X, Li X, Ding S. DNA-Guided Extracellular-Vesicle Metallization with High Catalytic Activity for Accurate Diagnosis of Pulmonary Nodules. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2208142. [PMID: 37066711 DOI: 10.1002/smll.202208142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/19/2023] [Indexed: 06/19/2023]
Abstract
Sensitive and specific analysis of extracellular vesicles (EVs) offers a promising minimally invasive way to identify malignant pulmonary nodules from benign lesions. However, accurate analysis of EVs is subject to free target proteins in blood samples, which compromises the clinical diagnosis value of EVs. Here a DNA-guided extracellular-vesicle metallization (DEVM) strategy is described for ultrasensitive and specific analysis of EV protein biomarkers and classification of pulmonary nodules. The facile DEVM process mainly includes the incorporation of DNA labeled with cholesterol and thiol groups into EV membranes and subsequent deposition of Au3+ and Pt4+ to allow the DNA-functionalized EVs to be encapsulated with AuPt nanoshells. It is found that the synthesized AuPt-metallized EVs possess extrinsic peroxidase-like activity. Utilizing the feature of the catalytic metal nanoshells just growth on the EV membranes, the DEVM method enables multiparametric recognition of target proteins and EV membranes and can produce an amplified colorimetric signal, avoiding the interference of free proteins. By profiling four surface proteins of EVs from 48 patients with pulmonary nodules, the highest area under the receiver operating characteristic curve (0.9983) is obtained. Therefore, this work provides a feasible EVs analysis tool for accurate pulmonary nodules management.
Collapse
Affiliation(s)
- Yirong Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Songzhi Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jun Lu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Dandan Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Haiping Wu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Lu Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xinyu Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xin Gao
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yuan Xu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Zijie Zeng
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Li Zeng
- Department of Laboratory Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, Chongqing, 400016, China
| | - Xiaojuan Ding
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xinmin Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
20
|
Borm FJ, Smit EF. Poziotinib for HER2 Exon 20-Mutated NSCLC: Addition or Burden to the Therapeutic Arsenal? J Thorac Oncol 2023; 18:964-966. [PMID: 37479324 DOI: 10.1016/j.jtho.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 07/23/2023]
Affiliation(s)
- Frank J Borm
- Department of Pulmonary Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Egbert F Smit
- Department of Pulmonary Diseases, Leiden University Medical Centre, Leiden, The Netherlands; Department of Thoracic Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek hospital (NKI-AvL), Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Sentana-Lledo D, Academia E, Viray H, Rangachari D, Kobayashi SS, VanderLaan PA, Costa DB. EGFR exon 20 insertion mutations and ERBB2 mutations in lung cancer: a narrative review on approved targeted therapies from oral kinase inhibitors to antibody-drug conjugates. Transl Lung Cancer Res 2023; 12:1590-1610. [PMID: 37577308 PMCID: PMC10413034 DOI: 10.21037/tlcr-23-98] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/14/2023] [Indexed: 08/15/2023]
Abstract
Background and Objective This review will provide an overview of EGFR and ERBB2 mutations in non-small-cell lung cancer (NSCLC) with a focus on recent clinical approvals. Methods We obtained data from the literature in accordance with narrative review reporting guidelines. Key Content and Findings EGFR mutations are present in up to 15-20% of all NSCLCs; amongst these, 10% correspond to kinase domain insertions in exon 20. Structurally similar, ERBB2 (HER2) mutations occurs in 1-4% of NSCLCs, mostly consisting of insertions or point mutations. The majority of EGFR exon 20 insertions occur within the loop following the regulatory C-helix and activate the kinase domain of EGFR without generating a therapeutic window to gefitinib, erlotinib, afatinib, dacomitinib or osimertinib. Mobocertinib represents a novel class of covalent EGFR inhibitors with a modest therapeutic window to these mutants and induces anti-tumor responses in a portion of patients [at 160 mg/day: response rate of <30% with duration of response (DoR) >17 months and progression-free survival (PFS) of >7 months] albeit with mucocutaneous and gastrointestinal toxicities. The bi-specific EGFR-MET antibody amivantamab-vmjw has modest but broad preclinical activity in EGFR-driven cancers and specifically for EGFR exon 20 insertion-mutated NSCLC has response rates <40% and PFS of <8.5 months at the cost of both infusion-related plus on-target toxicities. Both drugs were approved in 2021. The clinical development of kinase inhibitors for ERBB2-mutated NSCLC has been thwarted by mucocutaneous/gastrointestinal toxicities that preclude a pathway for drug approval, as the case of poziotinib. However, the activation of ERBB2 has allowed for repurposing of antibody-drug conjugates (ADCs) that target ERBB2 with cytotoxic payloads. The FDA approved fam-trastuzumab deruxtecan-nxki in 2022 for NSCLC based on response rate of >55%, DoR >9 months, PFS >8 months and manageable adverse events (including cytopenias, nausea and less commonly pneumonitis). Other therapies in clinical development include sunvozertinib and zipalertinib, among others. In addition, traditional cytotoxic chemotherapy has some activity in these tumors. Conclusions The approvals of mobocertinib, amivantamab, and trastuzumab deruxtecan represent the first examples of precision oncology for EGFR exon 20 insertion-mutated and ERBB2-mutated NSCLCs.
Collapse
Affiliation(s)
- Daniel Sentana-Lledo
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Emmeline Academia
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hollis Viray
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Deepa Rangachari
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Susumu S. Kobayashi
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Paul A. VanderLaan
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Daniel B. Costa
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Restrepo JC, Dueñas D, Corredor Z, Liscano Y. Advances in Genomic Data and Biomarkers: Revolutionizing NSCLC Diagnosis and Treatment. Cancers (Basel) 2023; 15:3474. [PMID: 37444584 DOI: 10.3390/cancers15133474] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a significant public health concern with high mortality rates. Recent advancements in genomic data, bioinformatics tools, and the utilization of biomarkers have improved the possibilities for early diagnosis, effective treatment, and follow-up in NSCLC. Biomarkers play a crucial role in precision medicine by providing measurable indicators of disease characteristics, enabling tailored treatment strategies. The integration of big data and artificial intelligence (AI) further enhances the potential for personalized medicine through advanced biomarker analysis. However, challenges remain in the impact of new biomarkers on mortality and treatment efficacy due to limited evidence. Data analysis, interpretation, and the adoption of precision medicine approaches in clinical practice pose additional challenges and emphasize the integration of biomarkers with advanced technologies such as genomic data analysis and artificial intelligence (AI), which enhance the potential of precision medicine in NSCLC. Despite these obstacles, the integration of biomarkers into precision medicine has shown promising results in NSCLC, improving patient outcomes and enabling targeted therapies. Continued research and advancements in biomarker discovery, utilization, and evidence generation are necessary to overcome these challenges and further enhance the efficacy of precision medicine. Addressing these obstacles will contribute to the continued improvement of patient outcomes in non-small cell lung cancer.
Collapse
Affiliation(s)
- Juan Carlos Restrepo
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
| | - Diana Dueñas
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
| | - Zuray Corredor
- Grupo de Investigaciones en Odontología (GIOD), Facultad de Odontología, Universidad Cooperativa de Colombia, Pasto 520002, Colombia
- Facultad de Salud, Departamento de Ciencias Básicas, Universidad Libre, Cali 760026, Colombia
| | - Yamil Liscano
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
| |
Collapse
|
23
|
Grodzka A, Knopik-Skrocka A, Kowalska K, Kurzawa P, Krzyzaniak M, Stencel K, Bryl M. Molecular alterations of driver genes in non-small cell lung cancer: from diagnostics to targeted therapy. EXCLI JOURNAL 2023; 22:415-432. [PMID: 37346803 PMCID: PMC10279966 DOI: 10.17179/excli2023-6122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023]
Abstract
Lung cancer is the leading cause of cancer death all over the world. The majority (80-85 %) of lung cancer cases are classified as non-small cell lung cancer (NSCLC). Within NSCLC, adenocarcinoma (AC) and squamous cell carcinoma (SCC) are the most often recognized. The histological and immunohistochemical examination of NSCLC is a basic diagnostic tool, but insufficient for comprehensive therapeutic decisions. In some NSCLC patients, mainly adenocarcinoma, molecular alterations in driver genes, like EGFR, KRAS, HER2, ALK, MET, BRAF, RET, ROS1, and NTRK are recognized. The frequency of some of those changes is different depending on race, and between smokers and non-smokers. The molecular diagnostics of NSCLC using modern methods, like next-generation sequencing, is essential in estimating targeted, personalized therapy. In recent years, a breakthrough in understanding the importance of molecular studies for the precise treatment of NSCLC has been observed. Many new drugs were approved, including tyrosine kinase and immune checkpoint inhibitors. Clinical trials testing novel molecules like miRNAs and trials with CAR-T cells (chimeric antigen receptor - T cells) dedicated to NSCLC patients are ongoing.
Collapse
Affiliation(s)
- Anna Grodzka
- Department of Cell Biology, Faculty of Biology, Adam Mickiewicz University of Poznan, Poland
| | | | - Katarzyna Kowalska
- Department of Oncological Pathology, University Clinical Hospital in Poznan, Poznan University of Medical Sciences, Poland
| | - Pawel Kurzawa
- Department of Oncological Pathology, University Clinical Hospital in Poznan, Poznan University of Medical Sciences, Poland
- Department of Clinical Pathology and Immunology, Poznan University of Medical Sciences, Poland
| | - Monika Krzyzaniak
- Department of Oncological Pathology, University Clinical Hospital in Poznan, Poznan University of Medical Sciences, Poland
| | - Katarzyna Stencel
- Department of Clinical Oncology with the Subdepartment of Diurnal Chemotherapy, E. J. Zeyland Wielkopolska Center of Pulmonology and Thoracic Surgery, Poznan, Poland
| | - Maciej Bryl
- Department of Clinical Oncology with the Subdepartment of Diurnal Chemotherapy, E. J. Zeyland Wielkopolska Center of Pulmonology and Thoracic Surgery, Poznan, Poland
| |
Collapse
|
24
|
Kargbo RB. Effective Combination Therapies for the Treatment of HER2 Cancer. ACS Med Chem Lett 2023; 14:231-232. [PMID: 36923923 PMCID: PMC10009784 DOI: 10.1021/acsmedchemlett.2c00544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Indexed: 02/09/2023] Open
Abstract
Breast cancer (BC) is the primary cause of cancer-related death among women worldwide. The human epidermal growth factor receptor type 2 (HER2)-positive BC accounts for ∼15% of all BCs and a relatively poor prognosis. The disclosure in this patent highlight relates to a combination therapy comprising inavolisib (GDC-0077) and a HER2-targeted therapy such as trastuzumab, pertuzumab, or their combination for the treatment of HER2-overexpressing breast cancer.
Collapse
|
25
|
Wang Z, Xing Y, Li B, Li X, Liu B, Wang Y. Molecular pathways, resistance mechanisms and targeted interventions in non-small-cell lung cancer. MOLECULAR BIOMEDICINE 2022; 3:42. [PMID: 36508072 PMCID: PMC9743956 DOI: 10.1186/s43556-022-00107-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/03/2022] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. The discovery of tyrosine kinase inhibitors effectively targeting EGFR mutations in lung cancer patients in 2004 represented the beginning of the precision medicine era for this refractory disease. This great progress benefits from the identification of driver gene mutations, and after that, conventional and new technologies such as NGS further illustrated part of the complex molecular pathways of NSCLC. More targetable driver gene mutation identification in NSCLC patients greatly promoted the development of targeted therapy and provided great help for patient outcomes including significantly improved survival time and quality of life. Herein, we review the literature and ongoing clinical trials of NSCLC targeted therapy to address the molecular pathways and targeted intervention progress in NSCLC. In addition, the mutations in EGFR gene, ALK rearrangements, and KRAS mutations in the main sections, and the less common molecular alterations in MET, HER2, BRAF, ROS1, RET, and NTRK are discussed. The main resistance mechanisms of each targeted oncogene are highlighted to demonstrate the current dilemma of targeted therapy in NSCLC. Moreover, we discuss potential therapies to overcome the challenges of drug resistance. In this review, we manage to display the current landscape of targetable therapeutic patterns in NSCLC in this era of precision medicine.
Collapse
Affiliation(s)
- Zixi Wang
- grid.412901.f0000 0004 1770 1022Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Yurou Xing
- grid.412901.f0000 0004 1770 1022Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Bingjie Li
- grid.412901.f0000 0004 1770 1022Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Xiaoyu Li
- grid.412901.f0000 0004 1770 1022Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan China ,grid.412901.f0000 0004 1770 1022State Key Laboratory Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Bin Liu
- grid.54549.390000 0004 0369 4060Department of Medical Oncology, School of Medicine, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, Sichuan China
| | - Yongsheng Wang
- grid.412901.f0000 0004 1770 1022Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China ,grid.412901.f0000 0004 1770 1022State Key Laboratory Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| |
Collapse
|