1
|
Wang L, Wen S, Zhu W, Zhang Z, Cheng Y. Response to trastuzumab deruxtecan and delayed immune-related events in a patient with metastatic HER2-positive NSCLC: a case report and literature review. Front Oncol 2025; 14:1469438. [PMID: 39896189 PMCID: PMC11782023 DOI: 10.3389/fonc.2024.1469438] [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: 07/23/2024] [Accepted: 12/24/2024] [Indexed: 02/04/2025] Open
Abstract
Trastuzumab deruxtecan (DS-8201) is an antibody-drug conjugate (ADC) designed to target HER2 mutations. We reported a case study demonstrating a favorable response to DS-8201 in a patient with HER2 mutation-positive non-small cell lung cancer (NSCLC) who exhibited resistance to initial immunotherapy, along with delayed immune-related events of hypoadrenocorticism occurring five months after discontinuation of immune checkpoint inhibitors. After reviewing the relevant literature, we discussed the mechanism of ADC agents underlying their anti-tumor activity and the potential impact of DS-8201 on the tumor microenvironment. This case highlights the efficacy of DS-8201 in NSCLC, particularly in individuals who have failed first-line immunotherapy, and provide valuable insights for clinicians exploring innovative strategies for the management of patients with lung cancer.
Collapse
Affiliation(s)
- Lu Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shidi Wen
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wenjia Zhu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhiyang Zhang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Melosky B, Juergens RA, Banerji S, Sacher A, Wheatley-Price P, Snow S, Tsao MS, Leighl NB, Martins I, Cheema P, Liu G, Chu QSC. The continually evolving landscape of novel therapies in oncogene-driven advanced non-small-cell lung cancer. Ther Adv Med Oncol 2025; 17:17588359241308784. [PMID: 39776537 PMCID: PMC11705342 DOI: 10.1177/17588359241308784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a highly heterogeneous disease that is frequently associated with a host of known oncogenic alterations. Advances in molecular diagnostics and drug development have facilitated the targeting of novel alterations such that the majority of NSCLC patients have driver mutations that are now clinically actionable. The goal of this review is to gain insights into clinical research and development principles by summary, analysis, and discussion of data on agents targeting known alterations in oncogene-driven, advanced NSCLC beyond those in the epidermal growth factor receptor (EGFR) and the anaplastic lymphoma kinase (ALK). A search of published and presented literature was conducted to identify prospective trials and integrated analyses reporting outcomes for agents targeting driver gene alterations (except those in EGFR and ALK) in molecularly selected, advanced NSCLC. Clinical efficacy data were extracted from eligible reports and summarized in text and tables. Findings show that research into alteration-directed therapies in oncogene-driven, advanced NSCLC is an extremely active research field. Ongoing research focuses on the expansion of new agents targeting both previously identified targets (particularly hepatocyte growth factor receptor (MET), human epidermal growth factor receptor 2 (HER2), and Kirsten rat sarcoma viral oncogene homolog (KRAS)) as well as novel, potentially actionable targets (such as neuregulin-1 (NRG1) and phosphatidylinositol 3-kinase (PI3K)). The refinement of biomarker selection criteria and the development of more selective and potent agents are allowing for increasingly specific and effective therapies and the expansion of clinically actionable alterations. Clinical advances in this field have resulted in a large number of regulatory approvals over the last 3 years. Future developments should focus on the continued application of alteration therapy matching principles and the exploration of novel ways to target oncogene-driven NSCLC.
Collapse
Affiliation(s)
- Barbara Melosky
- Medical Oncology, BC Cancer Agency—Vancouver, University of British Columbia, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | | | - Shantanu Banerji
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Paul Wheatley-Price
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Stephanie Snow
- QEII Health Sciences Centre, Dalhousie University, Halifax, NS, Canada
| | - Ming-Sound Tsao
- University Health Network and Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Natasha B. Leighl
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | | | - Parneet Cheema
- William Osler Health System, University of Toronto, Brampton, ON, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Quincy S. C. Chu
- Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
3
|
Zhang M, Wang L, Wang Q, Yang J, Peng W, Li X, Shi M, Lu K. Efficacy of disitamab vedotin in non-small cell lung cancer with HER2 alterations: a multicenter, retrospective real-world study. Front Oncol 2024; 14:1441025. [PMID: 39568568 PMCID: PMC11576286 DOI: 10.3389/fonc.2024.1441025] [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: 05/30/2024] [Accepted: 09/16/2024] [Indexed: 11/22/2024] Open
Abstract
Background Non-small cell lung cancer (NSCLC) with human epidermal growth factor receptor 2 (HER2) alterations poses a substantial treatment challenge. Current HER2-targeted therapies offer limited efficacy. Antibody-drug conjugates (ADCs) targeting HER2 have emerged as a promising therapeutic strategy. This study aimed to evaluate the clinical response to a novel ADC drug Disitamab vedotin (RC48) in advanced NSCLC with HER2 alterations. Methods This study conducted a retrospective review of patients harboring HER2 alterations treated with RC48 in the real world. Clinical outcomes were evaluated in terms of objective response rate (ORR), disease control rate (DCR), and progression-free survival (PFS). Results Out of 22 patients, 21 (95.5%) received RC48 combination therapy, while one received RC48 monotherapy. The ORR of all patients reached 45.5%, and the DCR stood at 90.9%. The median PFS (mPFS) was 7.5 months. Among patients receiving RC48 combination therapy, the ORR was 47.7%, and the mPFS of 8.1 months. The combination of RC48 with platinum+/- bevacizumab resulted in the highest ORR of 71.4% (5 out of 7 patients), with HER2 TKI following at a 50.0% ORR (4 out of 8 patients). First-line (1L) treatment with RC48 showed an ORR of 62.5% (5 out of 8 patients), second-line (2L) treatments had a 57.1% ORR (4 out of 7 patients), and beyond second-line (>2L) treatments exhibited a 14.3% ORR (1 out of 7 patients). Patients with 1L, 2L, or >2L treatment had a mPFS of 8.1 months, 7.2 months, and 7.4 months, respectively. Patients with HER2 mutations or amplifications, and those with concurrent mutations and amplifications at baseline, showed mPFS of 8.1 months, 9.4 months, and 7.4 months, respectively. The mPFS was significantly longer in patients with HER2 amplification. The most common adverse events included hand-foot syndrome (54.5%), asthenia (50.0%), decreased white blood cell count (45.5%), and liver impairment (45.5%). Grade 3 adverse events occurred in one (4.5%) patient. Conclusion RC48, particularly in combination regimens, demonstrates promising efficacy in advanced NSCLC with HER2 alterations. These findings underscore the need for further research to validate RC48's application in clinical practice.
Collapse
Affiliation(s)
- Meiling Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Wang
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiu Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Peng
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoyou Li
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Meiqi Shi
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Kaihua Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Odintsov I, Makarem M, Nishino M, Bachert SE, Zhang T, LoPiccolo J, Paweletz CP, Gokhale PC, Ivanova E, Saldanha A, Rudin CM, Lockwood WW, Ladanyi M, Somwar R, Jänne PA, Sholl LM. Prevalence and Therapeutic Targeting of High-Level ERBB2 Amplification in NSCLC. J Thorac Oncol 2024; 19:732-748. [PMID: 38154514 PMCID: PMC11957432 DOI: 10.1016/j.jtho.2023.12.019] [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: 10/21/2023] [Revised: 12/12/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023]
Abstract
INTRODUCTION ERBB2 amplification in lung cancer remains poorly characterized. HER2 (encoded by ERBB2) is a transmembrane tyrosine kinase capable of ligand-independent dimerization and signaling when overexpressed, and a common cause of HER2 overexpression is ERBB2 amplification. Here, we evaluated the clinicopathologic and genomic characteristics of ERBB2-amplified NSCLC and explored a HER2 antibody-drug conjugate (ADC) therapeutic strategy. METHODS Our institutional next-generation DNA sequencing data (OncoPanel) from 5769 NSCLC samples (5075 patients) were queried for cases having high-level ERBB2 amplification (≥6 copies). Clinical and demographic characteristics were extracted from the electronic medical records. Efficacy of the pan-ERBB inhibitor afatinib or HER2 ADCs (trastuzumab deruxtecan and trastuzumab emtansine) was evaluated in NSCLC preclinical models and patients with ERBB2 amplification. RESULTS High-level ERBB2 amplification was identified in 0.9% of lung adenocarcinomas and reliably predicted overexpression of HER2. ERBB2 amplification events are detected in two distinct clinicopathologic and genomic subsets of NSCLC: as the sole mitogenic driver in tumors arising in patients with a smoking history or as a concomitant alteration with other mitogenic drivers in patients with a light or never smoking history. We further reveal that trastuzumab deruxtecan is effective therapy in in vitro and in vivo preclinical models of NSCLC harboring ERBB2 amplification and report two cases of clinical activity of an anti-HER2 ADC in patients who acquired ERBB2 amplification after previous targeted therapy. CONCLUSIONS High-level ERBB2 amplification reliably predicts HER2 overexpression in patients with NSCLC, and HER2 ADC is effective therapy in this population.
Collapse
Affiliation(s)
- Igor Odintsov
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maisam Makarem
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mizuki Nishino
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sara Emily Bachert
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | - Tom Zhang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; School of Medicine, New York Medical College, Valhalla, New York
| | - Jaclyn LoPiccolo
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Cloud P Paweletz
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Prafulla C Gokhale
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elena Ivanova
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Aisha Saldanha
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William W Lockwood
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Romel Somwar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
5
|
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
|
6
|
Loeffler E, Ancel J, Dalstein V, Deslée G, Polette M, Nawrocki-Raby B. HER2 Alterations in Non-Small Cell Lung Cancer: Biologico-Clinical Consequences and Interest in Therapeutic Strategies. Life (Basel) 2023; 14:64. [PMID: 38255679 PMCID: PMC10820545 DOI: 10.3390/life14010064] [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: 10/30/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Lung cancer stands as the first cause of death by cancer in the world. Despite the improvement in patients' outcomes in the past decades through the development of personalized medicine approaches, a substantial portion of patients remains ineligible for targeted therapies due to the lack of a "druggable" molecular target. HER2, a receptor tyrosine kinase member of the EGFR/ErbB family, is known to show oncogenic properties. In this review, we focus on the different HER2 dysregulation mechanisms that have been observed in non-small cell lung cancer (NSCLC): gene mutation, gene amplification, protein overexpression and protein hyper-phosphorylation, the latter suggesting that HER2 dysregulation can occur independently of any molecular aberration. These HER2 alterations inevitably have consequences on tumor biology. Here, we discuss how they are not only involved in abnormal proliferation and survival of cancer cells but also potentially in increased angiogenic properties, mesenchymal features and tumor immune escape. Finally, we review the impact of these HER2 alterations in various therapeutic approaches. While standard chemotherapy and groundbreaking immunotherapy seem rather ineffective for HER2-altered NSCLCs, the development of HER2-targeted therapies such as tyrosine kinase inhibitors, anti-HER2 antibodies and especially antibody-drug conjugates could provide new hopes for patients.
Collapse
Affiliation(s)
- Emma Loeffler
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
| | - Julien Ancel
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
- CHU de Reims, Hôpital Maison-Blanche, Service de Pneumologie, 51092 Reims, France
| | - Véronique Dalstein
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
- CHU de Reims, Pôle de Biologie Territoriale, Service de Pathologie, 51092 Reims, France
| | - Gaëtan Deslée
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
- CHU de Reims, Hôpital Maison-Blanche, Service de Pneumologie, 51092 Reims, France
| | - Myriam Polette
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
- CHU de Reims, Pôle de Biologie Territoriale, Service de Pathologie, 51092 Reims, France
| | - Béatrice Nawrocki-Raby
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
| |
Collapse
|
7
|
Nützinger J, Bum Lee J, Li Low J, Ling Chia P, Talisa Wijaya S, Chul Cho B, Min Lim S, Soo RA. Management of HER2 alterations in non-small cell lung cancer - The past, present, and future. Lung Cancer 2023; 186:107385. [PMID: 37813015 DOI: 10.1016/j.lungcan.2023.107385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/11/2023]
Abstract
HER2 mutations, which account for 2-4% of non-small cell lung cancer (NSCLC), are distinct molecular alterations identified via next generation sequencing (NGS). Previously, treatment outcomes in HER2-mutant metastatic NSCLC were dismal, showing limited clinical benefit with platinum-based chemotherapy with or without immunotherapy. In contrast to HER2-altered breast and gastric cancer, HER2-mutant NSCLC does not benefit from HER2 targeting agents such as trastuzumab or TDM1. HER2 mutations are also inherently different from HER2 overexpression and amplification. Currently, trastuzumab deruxtecan, a HER2 targeting antibody drug conjugate (ADC) is the first and only approved treatment option for patients with HER2-mutant metastatic NSCLC after failure with standard treatment. In this review, we summarized the biology of HER2 and detection of HER2 overexpression, amplification and mutations, as well as general landscape of landmark and ongoing clinical trials encompassing from chemotherapy to targeted agents, including tyrosine kinase inhibitors (TKIs), ADCs and investigational agents.
Collapse
Affiliation(s)
- Jorn Nützinger
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jia Li Low
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Puey Ling Chia
- Department of Medical Oncology, Tan Tock Seng Hospital, Singapore
| | | | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Ross A Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore.
| |
Collapse
|
8
|
Uy NF, Merkhofer CM, Baik CS. HER2 in Non-Small Cell Lung Cancer: A Review of Emerging Therapies. Cancers (Basel) 2022; 14:cancers14174155. [PMID: 36077691 PMCID: PMC9454740 DOI: 10.3390/cancers14174155] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/03/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary There are growing data on targeting HER2 alterations, which include gene mutations, gene amplifications, and protein overexpression, for non-small cell lung cancer (NSCLC). Currently, there are limited targeted therapies approved for NSCLC patients with HER2 alterations, and this remains an unmet clinical need. There has been an influx of research on antibody–drug conjugates, monoclonal antibodies, and tyrosine kinase inhibitors. This review discusses the diagnostic challenges of HER2 alterations in NSCLC and summarizes recent progress in HER2 targeted drugs for both clinicians and researchers treating this patient population. Abstract Human epidermal growth factor receptor 2 (HER2), a member of the ERBB family of tyrosine kinase receptors, has emerged as a therapeutic target of interest for non-small cell lung cancer (NSCLC) in recent years. Activating HER2 alterations in NSCLC include gene mutations, gene amplifications, and protein overexpression. In particular, the HER2 exon 20 mutation is now a well clinically validated biomarker. Currently, there are limited targeted therapies approved for NSCLC patients with HER2 alterations. This remains an unmet clinical need, as HER2 alterations are present in 7–27% of de novo NSCLC and may serve as a resistance mechanism in up to 10% of EGFR mutated NSCLC. There has been an influx of research on antibody–drug conjugates (ADCs), monoclonal antibodies, and tyrosine kinase inhibitors (TKIs) with mixed results. The most promising therapies are ADCs (trastuzumab-deruxtecan) and novel TKIs targeting exon 20 mutations (poziotinib, mobocertinib and pyrotinib); both have resulted in meaningful anti-tumor efficacy in HER2 mutated NSCLC. Future studies on HER2 targeted therapy will need to define the specific HER2 alteration to better select patients who will benefit, particularly for HER2 amplification and overexpression. Given the variety of HER2 targeted drugs, sequencing of these agents and optimizing combination therapies will depend on more mature efficacy data from clinical trials and toxicity profiles. This review highlights the challenges of diagnosing HER2 alterations, summarizes recent progress in novel HER2-targeted agents, and projects next steps in advancing treatment for the thousands of patients with HER2 altered NSCLC.
Collapse
|
9
|
NSCLC as the Paradigm of Precision Medicine at Its Finest: The Rise of New Druggable Molecular Targets for Advanced Disease. Int J Mol Sci 2022; 23:ijms23126748. [PMID: 35743191 PMCID: PMC9223783 DOI: 10.3390/ijms23126748] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 02/04/2023] Open
Abstract
Standard treatment for advanced non-small cell lung cancer (NSCLC) historically consisted of systemic cytotoxic chemotherapy until the early 2000s, when precision medicine led to a revolutionary change in the therapeutic scenario. The identification of oncogenic driver mutations in EGFR, ALK and ROS1 rearrangements identified a subset of patients who largely benefit from targeted agents. However, since the proportion of patients with druggable alterations represents a minority, the discovery of new potential driver mutations is still an urgent clinical need. We provide a comprehensive review of the emerging molecular targets in NSCLC and their applications in the advanced setting.
Collapse
|
10
|
Waliany S, Wakelee H, Ramchandran K, Das M, Huang J, Myall N, Li C, Pagtama J, Tisch AH, Neal JW. Characterization of ERBB2 (HER2) Alterations in Metastatic Non-small Cell Lung Cancer and Comparison of Outcomes of Different Trastuzumab-based Regimens. Clin Lung Cancer 2022; 23:498-509. [PMID: 35753988 DOI: 10.1016/j.cllc.2022.05.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022]
Abstract
INTRODUCTION About 3%-5% of mNSCLC have ERBB2 (HER2) alterations, but currently, there are no FDA-approved targeted therapies for this indication. We compared treatment response between trastuzumab-based and non-targeted regimens in ERBB2-mutant mNSCLC. METHODS This retrospective, single-institution study included patients with mNSCLC with ERBB2 alterations identified by next-generation sequencing. Best overall response was determined using Response Evaluation Criteria in Solid Tumors 1.1. RESULTS We identified 3 groups of patients: ERBB2-mutant/EGFR-wildtype mNSCLC (n = 33), ERBB2-amplified/EGFR-wildtype mNSCLC without concurrent ERBB2 mutations (n = 6), and ERBB2-altered/EGFR-mutant mNSCLC (n = 8). Observed mutations included A775_G776insYVMA (n = 23), Gly778_Pro780dup (n = 4), Ser310Phe (n = 3), and others (n = 5). Among the 33 with ERBB2-mutant/EGFR-wildtype mNSCLC, those with and without A775_G776insYVMA had significantly different median overall survival (OS) of 17.7 and 52.9 months, respectively (Cox regression multivariable HR: 5.03, 95% CI: 1.37-18.51, P = .02). In those with mNSCLC with A775_G776insYVMA, trastuzumab-based therapies were associated with greater OS (20.3 vs. 9.8 months; multivariable HR: 0.19, 95% CI: 0.04-0.87, P = .032). Objective response and disease control rates (median tumor size change) in the 33 patients with ERBB2-mutant/EGFR-wildtype mNSCLC were 40.0% and 80.0% (-35.8%), respectively, for patients treated with trastuzumab deruxtecan; 0% and 30.0% (-5.2%) for trastuzumab emtansine; and 7.1% and 50.0% (-13.0%) for trastuzumab/chemotherapy combinations. CONCLUSION In ERBB2-mutant/EGFR-wildtype mNSCLC, while most trastuzumab-based regimens had modest activity in this real-world analysis, trastuzumab deruxtecan had highest response rates and best tumor size reduction. Receipt of any trastuzumab-based regimen was associated with greater OS with A775_G776insYVMA. There remains an unmet need for approved targeted therapies for ERBB2-mutant/EGFR-wildtype NSCLC.
Collapse
Affiliation(s)
- Sarah Waliany
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Heather Wakelee
- Department of Medicine, Stanford University School of Medicine, Stanford, CA; Division of Oncology, Stanford University School of Medicine, Stanford, CA; Stanford Cancer Institute, Stanford, CA
| | - Kavitha Ramchandran
- Department of Medicine, Stanford University School of Medicine, Stanford, CA; Division of Oncology, Stanford University School of Medicine, Stanford, CA; Stanford Cancer Institute, Stanford, CA
| | - Millie Das
- Department of Medicine, Stanford University School of Medicine, Stanford, CA; Division of Oncology, Stanford University School of Medicine, Stanford, CA; Stanford Cancer Institute, Stanford, CA; Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA
| | - Jane Huang
- Department of Medicine, Stanford University School of Medicine, Stanford, CA; Division of Oncology, Stanford University School of Medicine, Stanford, CA; Stanford Cancer Institute, Stanford, CA
| | - Nathaniel Myall
- Department of Medicine, Stanford University School of Medicine, Stanford, CA; Division of Oncology, Stanford University School of Medicine, Stanford, CA; Stanford Cancer Institute, Stanford, CA
| | - Connie Li
- Stanford Cancer Institute, Stanford, CA
| | | | | | - Joel W Neal
- Department of Medicine, Stanford University School of Medicine, Stanford, CA; Division of Oncology, Stanford University School of Medicine, Stanford, CA; Stanford Cancer Institute, Stanford, CA.
| |
Collapse
|
11
|
Li Y, Wu X, Fang J, Zhao Q, Huang Y, Jiang L. Evaluation of fluorine-18-fluorodeoxyglucose PET/computed tomography and human epithelial growth factor receptor 2 expression in treatment-naive patients with lung adenocarcinoma. Nucl Med Commun 2022; 43:442-450. [PMID: 35045546 DOI: 10.1097/mnm.0000000000001530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Human epithelial growth factor receptor 2 (HER2) is overexpressed in several types of cancers. The correlation between tumor glucose activity and HER2 expression can vary. This study is a retrospective investigation of fluorine-18-fluorodeoxyglucose PET/computed tomography (18F-FDG PET/CT) and HER2 expression status in patients with lung adenocarcinoma. METHODS The maximum standard uptake value (SUVmax) of 18F-FDG PET/CT was compared with the HER2 expression status in pretreated patients with lung adenocarcinoma. Moreover, clinicopathological characteristics, including age, gender, smoking, serum tumor markers, tumor location, size, stage and genetic mutation, were also evaluated in groups with different HER2 expressions. Patients' progression-free survival (PFS) and overall survival (OS) were also analyzed. RESULTS Ninety-six patients with HER2 expression, including 54 patients with HER2 overexpression and 30 patients without HER2 expression were enrolled in this study. The primary pulmonary lesion was single in all patients, and all lesions were FDG-avid on PET/CT. SUVmax had no significant association with HER2 expression or overexpression in lung adenocarcinoma. Moreover, elevated serum CYFRA211 levels were obviously associated with HER2 expression but not associated with HER2 overexpression. There were no significant differences in other clinicopathological characteristics in groups with different HER2 expressions. Furthermore, multivariate Cox regression analysis revealed that SUVmax, HER2 expression and tumor node metastasis stage were independent predictors of PFS, and SUVmax, CYFRA211 and epidermal growth factor receptor mutation were independent predictors of OS. CONCLUSION SUVmax had no significant association with the HER2 expression status in lung adenocarcinoma. 18F-FDG PET/CT and HER2 expression could provide valuable prognostic information for treatment-naive patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Yuan Li
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai
| | - Xiaodong Wu
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai
| | - Juanjuan Fang
- Department of Radiology, Dezhou Second People's Hospital, Shandong Province
| | - Qingping Zhao
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai
| | - Yan Huang
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai
| | - Lei Jiang
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
12
|
Wu HX, Zhuo KQ, Wang K. Efficacy of targeted therapy in patients with HER2-positive non-small cell lung cancer: A systematic review and meta-analysis. Br J Clin Pharmacol 2021; 88:2019-2034. [PMID: 34820879 PMCID: PMC9302639 DOI: 10.1111/bcp.15155] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/09/2021] [Accepted: 11/09/2021] [Indexed: 02/05/2023] Open
Abstract
Anti-human epidermal growth factor receptor 2 (HER2) therapy is an effective treatment for HER2-positive gastric and breast malignancies. However, the efficacy of HER2-targeted therapy in non-small cell lung cancer (NSCLC) patients with HER2 alterations remains controversial. We searched studies on HER2-targeted therapy in NSCLC patients that reported objective response rate (ORR), disease control rate (DCR) and progressionfree survival (PFS) published from database inception to 30 May 2021. A total of 32 trials involving 958 patients were included. The ORRs of HER2-TKIs targeted therapy, humanised monoclonal antibody, trastuzumab-based treatment and antibody-drug conjugate (ADC) (T-DM1) were 22% (95% CI 11-31), 23% (95% CI 20-65), 26% (95% CI 14-39) and 16% (95% CI _6-37), while that of ADC (DS-8201) was 60% (95% CI 35-85). The DCRs of these groups were 59% (95% CI 49-69), 39% (95% CI _9-88), 63% (95% CI 37-89), 31% (95% CI 4-58) and 87% (95% CI 62-112), respectively. In the subgroup analysis, numerically higher ORRs and DCRs were observed in the poziotinib (38%; 75%) and pyrotinib (35%; 83%) groups. The median PFSs of these groups were 5.51 months, 3.09 months, 4.61 months, 2.65 months and 12.04 months, respectively. HER2-targeted therapy can be considered an acceptable treatment strategy for NSCLC patients with HER2 alterations. In particular, ADC (DS-8201), pyrotinib and poziotinib demonstrated promising anti-tumour activity in HER2-positive NSCLC.
Collapse
Affiliation(s)
- Hong-Xia Wu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Kai-Quan Zhuo
- Department of Neurosurgery, Suining Municipal Hospital of TCM, Suining, China
| | - Ke Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Azar I, Alkassis S, Fukui J, Alsawah F, Fedak K, Al Hallak MN, Sukari A, Nagasaka M. Spotlight on Trastuzumab Deruxtecan (DS-8201,T-DXd) for HER2 Mutation Positive Non-Small Cell Lung Cancer. LUNG CANCER-TARGETS AND THERAPY 2021; 12:103-114. [PMID: 34675733 PMCID: PMC8507417 DOI: 10.2147/lctt.s307324] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/17/2021] [Indexed: 12/04/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a proto-oncogene that, when mutated or overexpressed, plays an important role in oncogenesis. The landscape of HER2-positive breast cancer has changed dramatically over the past 2 decades with the FDA approval of a growing number of agents (antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates) targeting the HER2 receptor. HER2 inhibition has also been approved for HER2-positive gastric cancer. HER2 is amplified in 9% and mutated in 3% of lung cancer. Historically, HER2-targeted therapy for lung cancer with trastuzumab, pertuzumab, and trastuzumab emtansine has failed to demonstrate a survival benefit. Trastuzumab deruxtecan (T-DXd) is a novel antibody–drug conjugate with a tetrapeptide linker, which delivers a topoisomerase I inhibitor with a drug-to-antibody ratio of 7~8. The potency of the active payload, as well as its significant bystander effect, resulted in significant anti-tumor activity. The DESTINY-Lung01 trial evaluated T-DXd in HER2-positive non-squamous non-small cell lung cancer (NSCLC) and reported a progression-free survival of 14 months in HER2-mutated NSCLC, earning its breakthrough designation by the FDA. In this review, we will discuss the structural characteristics, pharmacodynamics, and pharmacokinetics of T-DXd. We will also shed light on the preclinical and ongoing clinical trials of T-DXd along with future directions in the management of HER2 positive lung cancer.
Collapse
Affiliation(s)
- Ibrahim Azar
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - Samer Alkassis
- Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jami Fukui
- University of Hawaii Manoa Cancer Center, John A. Burns School of Medicine, Honolulu, HI, USA
| | - Fares Alsawah
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - Kalub Fedak
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - Mohammed Najeeb Al Hallak
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - Ammar Sukari
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - Misako Nagasaka
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA.,Division of Neurology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| |
Collapse
|
14
|
Clinicopathologic features and treatment advances in cancers with HER2 alterations. Biochim Biophys Acta Rev Cancer 2021; 1876:188605. [PMID: 34358635 DOI: 10.1016/j.bbcan.2021.188605] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022]
Abstract
HER2 is one of the most important proteins of the epidermal growth factor receptor (EGFR) family, whose alterations include amplification, overexpression and gene mutation. Growing attention has been given to HER2 as a biomarker for prognosis, an indicator for treatment response and a target for new drugs. Tumors with HER2 alterations have been well studied in multiple locations as distinct entities for treatment, especially breast cancer, gastric cancer, lung cancer and colorectal cancer. These four cancers are the leading causes of cancer incidence and cancer-related death worldwide. The present study details the landscape of HER2 amplification/overexpression and mutations and gives an up-to-date analysis of current clinical trials in the four cancers mentioned above. Different HER2-altered cancers not only respond differently to HER2-targeting therapies but also display diverse survival outcomes. Even in the same type of cancer, HER2 amplification/overexpression differs from HER2 mutation in terms of clinicopathologic features and treatment strategies. As an emerging strategy in cancer treatment, immune checkpoint inhibitors demonstrate distinct outcomes in HER2-altered breast cancer, gastric cancer and lung cancer.
Collapse
|
15
|
Chakravarthi S, Karikalan B. Molecular Biomarkers for Lung Adenocarcinoma: A Short Review. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394716666200724164654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lung cancer is a disease with higher death rates and is responsible for around 2 million
deaths per year worldwide. Recently, several breakthroughs have been made in the field of lung
cancer that has led to a revolution in the management of lung cancer patients. Identification of
molecular markers and the implication of respective targeted therapies has been a great success in
the treatment of lung adenocarcinoma patients. Despite the fact that targeted therapy of lung adenocarcinomas
represents one of the significant milestones in the treatment of lung cancer that resulted
in increased survival rates even in advanced stages, the mortality rates of lung cancer still remain
to be significantly high. This warrants further research for gaining better insights into molecular alterations
that can lead to newer innovations in targeted drug therapy towards lung adenocarcinoma.
In this review, we briefly summarized the literature on molecular markers that are already in use.
We also consolidated newer molecular markers that are under study with the potential for being targeted
for therapies in future.
Collapse
|
16
|
Zeng J, Ma W, Young RB, Li T. Targeting HER2 genomic alterations in non-small cell lung cancer. JOURNAL OF THE NATIONAL CANCER CENTER 2021; 1:58-73. [PMID: 39035769 PMCID: PMC11256690 DOI: 10.1016/j.jncc.2021.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 12/28/2022] Open
Abstract
Oncogenic mutations and amplifications in the erythroblastic oncogene B (ERBB2), or human epidermal growth factor receptor 2 (HER2), have emerged as distinct oncogenic drivers and drug targets in non-small cell lung cancer (NSCLC). Each genomic alteration occurs in 2-4% of NSCLC by next generation sequencing and is associated with constitutive HER2 activation. The most common HER2 mutations in NSCLC are exon 20 mutation A775_G776insYVMA mutation in the kinase domain and S310F mutation in the extracellular domain. Unlike in breast and gastric cancer, HER2 protein overexpression in NSCLC is not validated to be a biomarker predictive of clinical response to HER2-targeted agents. High HER2 protein overexpression by immunohistochemistry (3+) only occurs in 2-4% of NSCLC. Until now HER2-targeted agents (such as afatinib and ado-trastuzumab emtansine) only demonstrate modest clinical activity in patients with HER2-mutant NSCLC. Retrospective studies show concern for inferior clinical benefit of immune checkpoint inhibitors in HER2-mutated NSCLC. Therefore, platinum-based chemotherapy with or without an anti-angiogenesis inhibitor remains the first line standard treatment for this patient population. In May 2020 trastuzumab deruxtecan (T-DXd) received the U.S. Food and Drug Administration breakthrough therapy designation for HER2-mutant metastatic NSCLC, and was added as an option for HER2-mutant NSCLC to the NCCN guidelines V1.2021. A global phase III study of pyrotinib compared to docetaxel as a second line therapy for advanced NSCLC harboring HER2 exon 20 mutations was just opened for enrollment in September 2020. In this review, we highlight the current knowledge and perspectives on targeting-HER2 genomic alterations in NSCLC.
Collapse
Affiliation(s)
- Jie Zeng
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Weijie Ma
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Richard Benjamin Young
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Tianhong Li
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| |
Collapse
|
17
|
Wu R, Yuan B, Li C, Wang Z, Song Y, Liu H. A narrative review of advances in treatment and survival prognosis of HER2-positive malignant lung cancers. J Thorac Dis 2021; 13:3708-3720. [PMID: 34277062 PMCID: PMC8264687 DOI: 10.21037/jtd-20-3265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2), as a receptor tyrosine kinase of EGF receptor family, whose mutation is often associated with even if less frequency but poor prognosis and shorter survival in pulmonary malignant tumor. HER2 status include mutation, overexpression, amplification and also some rare genotypes, detected by next generation sequencing (NGS), immunohistochemistry (IHC), and also fluorescence in situ hybridization (FISH). Different genotypes represent different therapeutic targets and indicate different clinical prognosis concluded by previous studies. Unfortunately, no standard guidelines for first-line treatment are widely recognized, and current therapeutic schedules include chemotherapy, radiotherapy, targeted therapy, and immunotherapy. Especially for patients with advanced metastasis, chemotherapy is based as a systemic therapy using studies of breast cancer or EGFR-positive lung adenocarcinoma as a template. Studies already explored treatment including EGFR tyrosine kinase inhibitors (TKIs) such as gefitinib and afatinib, and also trastuzumab and its conjugation like HER2-targeted antibody-drug conjugate trastuzumab emtansine (T-DM1) and conjugate trastuzumab deruxtecan (T-DXd). Also, he researches explored combination therapy with chemotherapy and TKIs or monoclonal antibodies. This review describes commonly used therapies for HER2-positive/HER2-overexpression patients and general relationship between genotypes of HER2, drug selection and final prognosis in order to provide suggestions for future diagnosis and treatment.
Collapse
Affiliation(s)
- Ranpu Wu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southeast University of Medicine, Nanjing, China
| | - Bingxiao Yuan
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing Medical University, Nanjing, China
| | - Chuling Li
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing Medical University, Nanjing, China
| | - Zimu Wang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southeast University of Medicine, Nanjing, China.,Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing Medical University, Nanjing, China
| | - Hongbing Liu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southeast University of Medicine, Nanjing, China.,Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Singh SS, Dahal A, Shrestha L, Jois SD. Genotype Driven Therapy for Non-Small Cell Lung Cancer: Resistance, Pan Inhibitors and Immunotherapy. Curr Med Chem 2020; 27:5274-5316. [PMID: 30854949 DOI: 10.2174/0929867326666190222183219] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/25/2019] [Accepted: 02/14/2019] [Indexed: 12/14/2022]
Abstract
Eighty-five percent of patients with lung cancer present with Non-small Cell Lung Cancer (NSCLC). Targeted therapy approaches are promising treatments for lung cancer. However, despite the development of targeted therapies using Tyrosine Kinase Inhibitors (TKI) as well as monoclonal antibodies, the five-year relative survival rate for lung cancer patients is still only 18%, and patients inevitably become resistant to therapy. Mutations in Kirsten Ras Sarcoma viral homolog (KRAS) and epidermal growth factor receptor (EGFR) are the two most common genetic events in lung adenocarcinoma; they account for 25% and 20% of cases, respectively. Anaplastic Lymphoma Kinase (ALK) is a transmembrane receptor tyrosine kinase, and ALK rearrangements are responsible for 3-7% of NSCLC, predominantly of the adenocarcinoma subtype, and occur in a mutually exclusive manner with KRAS and EGFR mutations. Among drug-resistant NSCLC patients, nearly half exhibit the T790M mutation in exon 20 of EGFR. This review focuses on some basic aspects of molecules involved in NSCLC, the development of resistance to treatments in NSCLC, and advances in lung cancer therapy in the past ten years. Some recent developments such as PD-1-PD-L1 checkpoint-based immunotherapy for NSCLC are also covered.
Collapse
Affiliation(s)
- Sitanshu S Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201, United States
| | - Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201, United States
| | - Leeza Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201, United States
| | - Seetharama D Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201, United States
| |
Collapse
|
19
|
Shrestha L, Singh SS, Parajuli P, Dahal A, Mattheolabakis G, Meyer S, Bhattacharjee J, Jois SD. In vivo studies of a peptidomimetic that targets EGFR dimerization in NSCLC. J Cancer 2020; 11:5982-5999. [PMID: 32922539 PMCID: PMC7477407 DOI: 10.7150/jca.46320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/17/2020] [Indexed: 12/25/2022] Open
Abstract
Studies related to lung cancer have shown a link between human epidermal growth factor receptor-2 (HER2) expression and poor prognosis in patients with non-small cell lung cancer (NSCLC). HER2 overexpression has been observed in 3-38% of NSCLC, while strong HER2 protein overexpression is found in 2.5% of NSCLC. However, HER2 dimerization is important in lung cancer, including EGFR mutated NSCLC. Since HER2 dimerization leads to cell proliferation, targeting the dimerization of HER2 will have a significant impact on cancer therapies. A peptidomimetic has been designed that can be used as a therapeutic agent for a subset of NSCLC patients overexpressing HER2 or possessing HER2 as well as EGFR mutation. A cyclic peptidomimetic (18) has been designed to inhibit protein-protein interactions of HER2 with its dimerization partners EGFR and HER3. Compound 18 exhibited antiproliferative activity in HER2-positive NSCLC cell lines at nanomolar concentrations. Western blot analysis showed that 18 inhibited phosphorylation of HER2 and Akt in vitro and in vivo. Stability studies of 18 at various temperature and pH (pH 1 and pH 7.6), and in the presence of liver microsomes indicated that 18 was stable against thermal and chemical degradation. Pharmacokinetic parameters were evaluated in nude mice by administrating single doses of 4 mg/kg and 6 mg/kg of 18 via IV. The anticancer activity of 18 was evaluated using an experimental metastasis lung cancer model in mice. Compound 18 suppressed the tumor growth in mice when compared to control. A proximity ligation assay further proved that 18 inhibits HER2:HER3 and EGFR: HER2 dimerization. Overall, these results suggest that 18 can be a potential treatment for HER2-dimerization related NSCLC.
Collapse
Affiliation(s)
- Leeza Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201
| | - Sitanshu S. Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201
| | - Pravin Parajuli
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201
| | - Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201
| | - Sharon Meyer
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201
| | - Joydeep Bhattacharjee
- Biology Program, School of Sciences, University of Louisiana, Monroe, Monroe, LA 71029
| | - Seetharama D. Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201
| |
Collapse
|
20
|
Zhou C, Li X, Wang Q, Gao G, Zhang Y, Chen J, Shu Y, Hu Y, Fan Y, Fang J, Chen G, Zhao J, He J, Wu F, Zou J, Zhu X, Lin X. Pyrotinib in HER2-Mutant Advanced Lung Adenocarcinoma After Platinum-Based Chemotherapy: A Multicenter, Open-Label, Single-Arm, Phase II Study. J Clin Oncol 2020; 38:2753-2761. [PMID: 32614698 DOI: 10.1200/jco.20.00297] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE Targeted therapies against non-small-cell lung cancer (NSCLC) harboring HER2 mutations remain an unmet need. In this study, we assessed the efficacy and safety of pyrotinib in patients with HER2-mutant advanced NSCLC in a prospective, multicenter, open-label, single-arm, phase II study. PATIENTS AND METHODS Patients with stage IIIB or IV HER2-mutant lung adenocarcinoma who were previously treated with platinum-based chemotherapy were enrolled to receive pyrotinib at a dose of 400 mg/d for 21-day cycles. The primary end point was objective response rate per independent review committee (IRC). RESULTS Between October 20, 2016, and December 10, 2018, 60 patients received pyrotinib monotherapy. At baseline, 58 (96.7%) were stage IV, and 25 (41.7%) received at least 2 lines of prior chemotherapy. As of data cutoff on June 20, 2019, IRC-assessed objective response rate was 30.0% (95% CI, 18.8% to 43.2%). All subgroups of patients with different HER2 mutation types showed a favorable objective response rate. The objective response rates were similar between patients with and without brain metastases (25.0% v 31.3%). The median duration of response was 6.9 months (95% CI, 4.9 to 11.1 months). The median progression-free survival was 6.9 months (95% CI, 5.5 to 8.3 months) per IRC. The median overall survival was 14.4 months (95% CI, 12.3 to 21.3 months). Treatment-related adverse events of grade 3 or 4 occurred in 28.3% of patients, with the most common being diarrhea (20.0%; all grade 3). No treatment-related deaths were reported. CONCLUSION Pyrotinib showed promising antitumor activity and an acceptable safety profile in chemotherapy-treated patients with HER2-mutant NSCLC.
Collapse
Affiliation(s)
- Caicun Zhou
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xingya Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Guanghui Gao
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | | | - Jianhua Chen
- Cancer Hospital of Central South University, Changsha, China
| | - Yongqian Shu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | - Yun Fan
- Zhejiang Cancer Hospital, Hangzhou, China
| | - Jian Fang
- Peking University Cancer Hospital and Institute, Beijing, China
| | - Gongyan Chen
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Jun Zhao
- Peking University Cancer Hospital and Institute, Beijing, China
| | - Jianxing He
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fengying Wu
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianjun Zou
- Jiangsu Hengrui Medicine Co, Ltd, Shanghai, China
| | - Xiaoyu Zhu
- Jiangsu Hengrui Medicine Co, Ltd, Shanghai, China
| | - Xiang Lin
- Jiangsu Hengrui Medicine Co, Ltd, Shanghai, China
| |
Collapse
|
21
|
Russo A, Lopes AR, McCusker MG, Garrigues SG, Ricciardi GR, Arensmeyer KE, Scilla KA, Mehra R, Rolfo C. New Targets in Lung Cancer (Excluding EGFR, ALK, ROS1). Curr Oncol Rep 2020; 22:48. [PMID: 32296961 DOI: 10.1007/s11912-020-00909-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Over the last two decades, the identification of targetable oncogene drivers has revolutionized the therapeutic landscape of non-small cell lung cancer (NSCLC). The extraordinary progresses made in molecular biology prompted the identification of several rare molecularly defined subgroups. In this review, we will focus on the novel and emerging actionable oncogenic drivers in NSCLC. RECENT FINDINGS Recently, novel oncogene drivers emerged as promising therapeutic targets besides the well-established EGFR mutations, and ALK/ROS1 rearrangements, considerably expanding the list of potential exploitable genetic aberrations. However, the therapeutic algorithm in these patients is far less defined. The identification of uncommon oncogene drivers is reshaping the diagnostic and therapeutic approach to NSCLC. The introduction of novel highly selective inhibitors is expanding the use of targeted therapies to rare and ultra-rare subsets of patients, further increasing the therapeutic armamentarium of advanced NSCLC.
Collapse
Affiliation(s)
- Alessandro Russo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA.,Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Contrada Papardo, 98158, Messina, Italy
| | - Ana Rita Lopes
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA.,Portuguese Institute of Oncology (IPO), Porto, Portugal
| | - Michael G McCusker
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Sandra Gimenez Garrigues
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Giuseppina R Ricciardi
- Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Contrada Papardo, 98158, Messina, Italy
| | - Katherine E Arensmeyer
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Katherine A Scilla
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Ranee Mehra
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA
| | - Christian Rolfo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S Greene Street Rm. N9E08, Baltimore, MD, 21201, USA.
| |
Collapse
|
22
|
Genova C, Rossi G, Tagliamento M, Rijavec E, Biello F, Cerbone L, Zullo L, Grossi F. Targeted therapy of oncogenic-driven advanced non-small cell lung cancer: recent advances and new perspectives. Expert Rev Respir Med 2020; 14:367-383. [DOI: 10.1080/17476348.2020.1714441] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Carlo Genova
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giovanni Rossi
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Italy
| | - Marco Tagliamento
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Erika Rijavec
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Federica Biello
- Oncology Unit, Ospedale Maggiore della Carità, Novara, Italy
| | - Luigi Cerbone
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lodovica Zullo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesco Grossi
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
23
|
Lorch G, Sivaprakasam K, Zismann V, Perdigones N, Contente-Cuomo T, Nazareno A, Facista S, Wong S, Drenner K, Liang WS, Amann JM, Sinicropi-Yao SL, Koenig MJ, La Perle K, Whitsett TG, Murtaza M, Trent JM, Carbone DP, Hendricks WPD. Identification of Recurrent Activating HER2 Mutations in Primary Canine Pulmonary Adenocarcinoma. Clin Cancer Res 2019; 25:5866-5877. [PMID: 31431454 DOI: 10.1158/1078-0432.ccr-19-1145] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/19/2019] [Accepted: 07/29/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Naturally occurring primary canine lung cancers share clinicopathologic features with human lung cancers in never-smokers, but the genetic underpinnings of canine lung cancer are unknown. We have charted the genomic landscape of canine lung cancer and performed functional characterization of novel, recurrent HER2 (ERBB2) mutations occurring in canine pulmonary adenocarcinoma (cPAC). EXPERIMENTAL DESIGN We performed multiplatform genomic sequencing of 88 primary canine lung tumors or cell lines. Additionally, in cPAC cell lines, we performed functional characterization of HER2 signaling and evaluated mutation-dependent HER2 inhibitor drug dose-response. RESULTS We discovered somatic, coding HER2 point mutations in 38% of cPACs (28/74), but none in adenosquamous (cPASC, 0/11) or squamous cell (cPSCC, 0/3) carcinomas. The majority (93%) of HER2 mutations were hotspot V659E transmembrane domain (TMD) mutations comparable to activating mutations at this same site in human cancer. Other HER2 mutations were located in the extracellular domain and TMD. HER2 V659E was detected in the plasma of 33% (2/6) of dogs with localized HER2 V659E tumors. HER2 V659E cPAC cell lines displayed constitutive phosphorylation of AKT and significantly higher sensitivity to the HER2 inhibitors lapatinib and neratinib relative to HER2-wild-type cell lines (IC50 < 200 nmol/L in HER2 V659E vs. IC50 > 2,500 nmol/L in HER2 WT). CONCLUSIONS This study creates a foundation for molecular understanding of and drug development for canine lung cancer. These data also establish molecular contexts for comparative studies in dogs and humans of low mutation burden, never-smoker lung cancer, and mutant HER2 function and inhibition.
Collapse
Affiliation(s)
- Gwendolen Lorch
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | | | | | | | | | | | | | - Shukmei Wong
- Translational Genomics Research Institute, Phoenix, Arizona
| | - Kevin Drenner
- Translational Genomics Research Institute, Phoenix, Arizona
| | - Winnie S Liang
- Translational Genomics Research Institute, Phoenix, Arizona
| | - Joseph M Amann
- Department of Internal Medicine, James Thoracic Center, The Ohio State University, Columbus, Ohio
| | - Sara L Sinicropi-Yao
- Department of Internal Medicine, James Thoracic Center, The Ohio State University, Columbus, Ohio
| | - Michael J Koenig
- Department of Internal Medicine, James Thoracic Center, The Ohio State University, Columbus, Ohio
| | - Krista La Perle
- Department of Veterinary Biosciences, Comparative Pathology and Mouse Phenotyping Shared Resource, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | | | | | | | - David P Carbone
- Department of Internal Medicine, James Thoracic Center, The Ohio State University, Columbus, Ohio
| | | |
Collapse
|
24
|
Subramanian J, Katta A, Masood A, Vudem DR, Kancha RK. Emergence of ERBB2 Mutation as a Biomarker and an Actionable Target in Solid Cancers. Oncologist 2019; 24:e1303-e1314. [PMID: 31292270 DOI: 10.1634/theoncologist.2018-0845] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 05/25/2019] [Indexed: 02/06/2023] Open
Abstract
The oncogenic role ERBB2 amplification is well established in breast and gastric cancers. This has led to the development of a well-known portfolio of monoclonal antibodies and kinase inhibitors targeting the ERBB2 kinase. More recently, activating mutations in the ERBB2 gene have been increasingly reported in multiple solid cancers and were shown to play an oncogenic role similar to that of ERBB2 amplification. Thus, ERBB2 mutations define a distinct molecular subtype of solid tumors and serve as actionable targets. However, efforts to target ERBB2 mutation has met with limited clinical success, possibly because of their low frequency, inadequate understanding of the biological activity of these mutations, and difficulty in separating the drivers from the passenger mutations. Given the current impetus to deliver molecularly targeted treatments for cancer, there is an important need to understand the therapeutic potential of ERBB2 mutations. Here we review the distribution of ERBB2 mutations in different tumor types, their potential as a novel biomarker that defines new subsets in many cancers, and current data on preclinical and clinical efforts to target these mutations. IMPLICATIONS FOR PRACTICE: A current trend in oncology is to identify novel genomic drivers of solid tumors and developing precision treatments that target them. ERBB2 amplification is an established therapeutic target in breast and gastric cancers, but efforts to translate this finding to other solid tumors with ERBB2 amplification have not been effective. Recently the focus has turned to targeting activating ERBB2 mutations. The year 2018 marked an important milestone in establishing ERBB2 mutation as an important actionable target in multiple cancer types. There have been several recent preclinical and clinical studies evaluating ERBB2 mutation as a therapeutic target with varying success. With increasing access to next-generation sequencing technologies in the clinic, oncologists are frequently identifying activating ERBB2 mutations in patients with cancer. There is a significant need both from the clinician and bench scientist perspectives to understand the current state of affairs for ERBB2 mutations.
Collapse
Affiliation(s)
- Janakiraman Subramanian
- Division of Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri, USA
- Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri, USA
| | - Archana Katta
- Molecular Medicine and Therapeutics Laboratory, Centre for Plant Molecular Biology, Osmania University, Hyderabad, India
| | - Ashiq Masood
- Division of Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri, USA
- Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri, USA
| | - Dashavantha Reddy Vudem
- Molecular Biology Laboratory, Centre for Plant Molecular Biology, Osmania University, Hyderabad, India
| | - Rama Krishna Kancha
- Molecular Medicine and Therapeutics Laboratory, Centre for Plant Molecular Biology, Osmania University, Hyderabad, India
| |
Collapse
|
25
|
Jiao H, Zhao X, Liu J, Ma T, Zhang Z, Zhang J, Wang J. In vivo imaging characterization and anticancer efficacy of a novel HER2 affibody and pemetrexed conjugate in lung cancer model. Nucl Med Biol 2018; 68-69:31-39. [PMID: 30578135 DOI: 10.1016/j.nucmedbio.2018.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/05/2018] [Accepted: 11/22/2018] [Indexed: 11/26/2022]
Abstract
INTRODUCTION In this study, a new agent consisting of HER2-specific affibody ZHER2:V2 and chemotherapy drug pemetrexed was synthesized to develop a new targeted drug. Its biological characteristics and anticancer efficacy were assessed in cells level and xenografts models by radiolabeling with technetium-99m. METHODS After the ZHER2:V2-pemetrexed conjugate was synthesized, radiolabeling of the conjugate was performed using its C-terminal 4 amino acids (Gly-Gly-Gly-Cys) as the chelating moiety. The radiochemical yield of the [99mTc]Tc-ZHER2:V2-pemetrexed was identified by instant thin-layer chromatography (ITLC). Stability of the radiolabeled conjugate was investigated both in vitro and in vivo. In vitro binding affinity and cell internalization study of the probe were performed in A549 cells (HER2-positive). Tumor uptake was evaluated by in vitro uptake assay in A549 cells and H23 cells (HER2-negative), and by in vivo biodistribution and SPECT imaging in A549 and H23 tumor-bearing mice. The antitumor efficacy of the ZHER2:V2-pemetrexed conjugate was evaluated in cells and xenograft models. RESULTS The ZHER2:V2-pemetrexed was successfully synthesized and conjugated with technetium-99 m, and acquired the radiochemical yield of 97.0 ± 0.3%. The stability of [99mTc]Tc-ZHER2:V2-pemetrexed was good in both physiological saline and human serum. The radiolabeled agent displayed excellent HER2-binding specificity and affinity in vitro, and was gradually internalized into the cells. Biodistribution study revealed obvious tumor uptake in A549 xenografts (percentage injected dose per gram, 2.6 ± 1.0%ID/g at 4 h postinjection), while the uptake in HER2-negative H23 tumors was much lower (0.2 ± 0.1%ID/g at 4 h postinjection, P < 0.01). SPECT imaging exhibited an intensity in the A549 xenograft which could be blocked by excess ZHER2:V2-pemetrexed. Treatment with ZHER2:V2-pemetrexed significantly impaired the tumor growth (P < 0.05), with less weight loss than pemetrexed. CONCLUSION [99mTc]Tc-ZHER2:V2-pemetrexed showed desirable property and HER2-specificity. The ZHER2:V2-pemetrexed conjugate could inhibit tumor growth of HER2-positive lung adenocarcinoma and may have the potential to become a targeted drug for lung cancer. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE The compound described herein performs HER2-targeting with favorable anticancer efficacy and offers the potential of novel targeting strategies for further tumor therapy.
Collapse
Affiliation(s)
- Honglei Jiao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| | - Jiahui Liu
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Tuo Ma
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Zhaoqi Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Jianfang Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| |
Collapse
|
26
|
Zhang P, Nie X, Wang B, Li L. Combined therapy with osimertinib and afatinib in a lung adenocarcinoma patient with EGFR T790M mutation and multiple HER2 alterations after resistance to icotinib: A case report. Thorac Cancer 2018; 9:1774-1777. [PMID: 30295016 PMCID: PMC6275817 DOI: 10.1111/1759-7714.12889] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/06/2018] [Accepted: 09/06/2018] [Indexed: 11/27/2022] Open
Abstract
Acquired resistance inevitably occurs after initial treatment with first-generation EGFR-tyrosine kinase inhibitors (TKIs). Several mechanisms have been identified, including EGFR T790M mutation and HER2 amplification. Herein, we present the case of a patient who progressed on first-generation EGFR-TKIs and developed EGFR T790M mutation, HER2 amplification, and HER2 mutation. The administration of single-agent osimertinib yielded an inconsistent response, with worsened pleural effusion and a reduction to lung metastases, but remarkably, a partial response was achieved after four weeks of treatment with combined osimertinib and afatinib, with grade 1 rash and grade 2 diarrhea. Our findings indicate an overlap of T790M, HER2 amplification, and HER2 mutation, which is rarely reported. Moreover, HER2 mutation was identified during the development of resistance, suggesting that HER2 mutation may cause resistance to first-generation EGFR-TKIs.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Xin Nie
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Bing Wang
- Department of In Vitro Diagnostic Testing, Beijing Institute of Medical Device Testing, Beijing, China
| | - Lin Li
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China
| |
Collapse
|
27
|
Oh IJ, Hur JY, Park CK, Kim YC, Kim SJ, Lee MK, Kim HJ, Lee KY, Lee JC, Choi CM. Clinical Activity of Pan-HER Inhibitors Against HER2-Mutant Lung Adenocarcinoma. Clin Lung Cancer 2018; 19:e775-e781. [DOI: 10.1016/j.cllc.2018.05.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 03/17/2018] [Accepted: 05/28/2018] [Indexed: 01/18/2023]
|
28
|
Li BT, Shen R, Buonocore D, Olah ZT, Ni A, Ginsberg MS, Ulaner GA, Offin M, Feldman D, Hembrough T, Cecchi F, Schwartz S, Pavlakis N, Clarke S, Won HH, Brzostowski EB, Riely GJ, Solit DB, Hyman DM, Drilon A, Rudin CM, Berger MF, Baselga J, Scaltriti M, Arcila ME, Kris MG. Ado-Trastuzumab Emtansine for Patients With HER2-Mutant Lung Cancers: Results From a Phase II Basket Trial. J Clin Oncol 2018; 36:2532-2537. [PMID: 29989854 PMCID: PMC6366814 DOI: 10.1200/jco.2018.77.9777] [Citation(s) in RCA: 394] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Purpose Human epidermal growth factor receptor 2 ( HER2, ERBB2)-activating mutations occur in 2% of lung cancers. We assessed the activity of ado-trastuzumab emtansine, a HER2-targeted antibody-drug conjugate, in a cohort of patients with HER2-mutant lung cancers as part of a phase II basket trial. Patients and Methods Patients received ado-trastuzumab emtansine at 3.6 mg/kg intravenously every 3 weeks until progression. The primary end point was overall response rate using Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. A Simon two-stage optimal design was used. Other end points included progression-free survival and toxicity. HER2 testing was performed on tumor tissue by next generation sequencing, fluorescence in situ hybridization, immunohistochemistry, and protein mass spectrometry. Results We treated 18 patients with advanced HER2-mutant lung adenocarcinomas. The median number of prior systemic therapies was two (range, zero to four prior therapies). The partial response rate was 44% (95% CI, 22% to 69%), meeting the primary end point. Responses were seen in patients with HER2 exon 20 insertions and point mutations in the kinase, transmembrane, and extracellular domains. Concurrent HER2 amplification was observed in two patients. HER2 immunohistochemistry ranged from 0 to 2+ and did not predict response, and responders had low HER2 protein expression measured by mass spectrometry. The median progression-free survival was 5 months (95% CI, 3 to 9 months). Toxicities included grade 1 or 2 infusion reactions, thrombocytopenia, and elevated hepatic transaminases. No patient stopped therapy as a result of toxicity or died on study. Conclusion Ado-trastuzumab emtansine is an active agent in patients with HER2-mutant lung cancers. This is the first positive trial in this molecular subset of lung cancers. Further use and study of this agent are warranted.
Collapse
Affiliation(s)
- Bob T. Li
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Ronglai Shen
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Darren Buonocore
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Zachary T. Olah
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Ai Ni
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Michelle S. Ginsberg
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Gary A. Ulaner
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Michael Offin
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Daniel Feldman
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Todd Hembrough
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Fabiola Cecchi
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Sarit Schwartz
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Nick Pavlakis
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Stephen Clarke
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Helen H. Won
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Edyta B. Brzostowski
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Gregory J. Riely
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - David B. Solit
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - David M. Hyman
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Alexander Drilon
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Charles M. Rudin
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Michael F. Berger
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - José Baselga
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Maurizio Scaltriti
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Maria E. Arcila
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| | - Mark G. Kris
- Bob T. Li, Ronglai Shen, Darren Buonocore, Zachary T. Olah, Ai Ni, Michelle S. Ginsberg, Gary A. Ulaner, Michael Offin, Daniel Feldman, Helen H. Won, Edyta B. Brzostowski, Gregory J. Riely, David B. Solit, David M. Hyman, Alexander Drilon, Charles M. Rudin, Michael F. Berger, José Baselga, Maurizio Scaltriti, Maria E. Arcila, and Mark G. Kris, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Bob T. Li, Nick Pavlakis, and Stephen Clarke, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; and Todd Hembrough, Fabiola Cecchi, and Sarit Schwartz, NantOmics, Rockville, MD
| |
Collapse
|
29
|
Peters S, Curioni-Fontecedro A, Nechushtan H, Shih JY, Liao WY, Gautschi O, Spataro V, Unk M, Yang JCH, Lorence RM, Carrière P, Cseh A, Chang GC. Activity of Afatinib in Heavily Pretreated Patients With ERBB2 Mutation-Positive Advanced NSCLC: Findings From a Global Named Patient Use Program. J Thorac Oncol 2018; 13:1897-1905. [PMID: 30096481 DOI: 10.1016/j.jtho.2018.07.093] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/03/2018] [Accepted: 07/31/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Approximately 1% to 4% of NSCLC tumors harbor erb-b2 receptor tyrosine kinase 2 (ERBB2) mutation; there is no approved targeted treatment for this subgroup. METHODS Patients with stage IV NSCLC that progressed after clinical benefit on erlotinib/gefitinib and/or had activating EGFR or ERBB2 mutations, had exhausted other treatments, and were ineligible for afatinib trials were enrolled in a named patient use program, receiving afatinib 30 to 50 mg/d on a compassionate basis within routine clinical practice. Efficacy and safety were retrospectively assessed in the subgroup with ERBB2 mutation-positive NSCLC. RESULTS Twenty-eight heavily pretreated patients in the named patient use program had a documented ERBB2 mutation by local testing. Median time-to-treatment failure (TTF; time from treatment initiation to discontinuation for any reason) was 2.9 months; eight patients (29%) had TTF greater than 1 year. Objective response rate was 19% (3 of 16 patients with response data achieved partial response) and disease control rate (DCR) was 69% (11 of 16). Among 12 patients for whom type of ERBB2 mutation was specified, 10 had a p.A775_G776insYVMA insertion in exon 20, four of whom (40%) remained on afatinib for more than 1 year. This subgroup had median TTF of 9.6 months, objective response rate of 33% (two of six), and disease control rate of 100% (six of six). CONCLUSIONS This analysis of patients treated in clinical practice provides further evidence of the activity of afatinib in ERBB2 mutation-positive NSCLC, and suggests that identification of specific subgroups with certain mutations, such as p.A775_G776ins/YVMA insertion in exon 20, could help optimize outcomes with ErbB2-targeted treatment.
Collapse
Affiliation(s)
- Solange Peters
- Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| | | | - Hovav Nechushtan
- Sharett Institute of Oncology, Hadassah Hebrew University Medical Center, Kiryat Hadassah, Jerusalem, Israel
| | - Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wei-Yu Liao
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Oliver Gautschi
- Medical Oncology, Cantonal Hospital Lucerne, Lucerne, Switzerland
| | - Vito Spataro
- Department of Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Mojca Unk
- Department for Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | | | - Robert M Lorence
- Oncology Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Philippe Carrière
- Risk Management Oncology, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Agnieszka Cseh
- Medical Department, Boehringer Ingelheim RCV GmbH & Co. KG, Vienna, Austria
| | - Gee-Chen Chang
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan; and the Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| |
Collapse
|
30
|
Trastuzumab and paclitaxel in patients with EGFR mutated NSCLC that express HER2 after progression on EGFR TKI treatment. Br J Cancer 2018; 119:558-564. [PMID: 30061586 PMCID: PMC6162232 DOI: 10.1038/s41416-018-0194-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/02/2018] [Accepted: 07/04/2018] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND HER2 expression and amplification are observed in ~15% of tumour biopsies from patients with a sensitising EGFR mutation who develop EGFR TKI resistance. It is unknown whether HER2 targeting in this setting can result in tumour responses. METHODS A single arm phase II study was performed to study the safety and efficacy of trastuzumab and paclitaxel treatment in patients with a sensitising EGFR mutation who show HER2 expression in a tumour biopsy (IHC ≥ 1) after progression on EGFR TKI treatment. Trastuzumab (first dose 4 mg/kg, thereafter 2 mg/kg) and paclitaxel (60 mg/m2) were dosed weekly until disease progression or unacceptable toxicity. The primary end-point was tumour response rate according to RECIST v1.1. RESULTS Twenty-four patients were enrolled. Nine patients were exon 21 L858R positive and fifteen exon 19 del positive. Median HER2 IHC was 2+ (range 1-3). For 21 patients, gene copy number by in situ hybridisation could be calculated: 5 copies/nucleus (n = 9), 5-10 copies (n = 8), and >10 copies (n = 4). An objective response was observed in 11/24 (46%) patients. Highest response rates were seen for patients with 3+ HER2 IHC (12 patients, ORR 67%) or HER2 copy number ≥10 (4 patients, ORR 100%). Median tumour change in size was 42% decrease (range -100% to +53%). Median duration of response was 5.6 (95% confidence interval [CI], 3.8 to 7.3) months. Treatment toxicity was mild with four patients experiencing grade ≥3 toxicity, including fatigue, neuropathy, neutropaenia, urinary tract infection, and pneumonitis. CONCLUSIONS Trastuzumab-paclitaxel induces objective tumour responses in 46% of EGFR TKI pretreated patients with an activating EGFR mutation and HER2 expression. The treatment was well tolerated. The relation between response rate and HER2 expression level and copy number suggests effective HER2 targeting by trastuzumab, although the combination with paclitaxel does not allow to determine the relative contribution of the individual drugs in terms of treatment efficacy.
Collapse
|
31
|
Zhao J, Chen X, Zheng J, Kong M, Wang B, Ding W. A genomic and clinicopathological study of non-small-cell lung cancers with discordant ROS1 gene status by fluorescence in-situ hybridisation and immunohistochemical analysis. Histopathology 2018; 73:19-28. [PMID: 29464758 DOI: 10.1111/his.13492] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 02/15/2018] [Indexed: 11/30/2022]
Abstract
AIMS ROS1 immunohistochemistry (IHC) using D4D6 antibody is a useful tool for screening patients with non-small-cell lung cancer (NSCLC) who may be suitable for targeted therapy. Many studies and our data have identified cases that express the ROS1 protein strongly but are negative for ROS1 by fluorescence in-situ hybridisation (FISH). The present study investigated the driver mutation and clinicopathological characteristics of 26 discordant cases (ROS1 IHC-positive but FISH-negative) to find new clues for distinguishing real ROS1-rearranged cases. METHODS AND RESULTS Tumours from 26 discordant cases were analysed for clinicopathological characteristics, mutations in EGFR, KRAS, ERBB2, BRAF and PIK3CA; fusions in ALK and RET; and amplifications in MET, ERBB2 and ROS1. ROS1-rearranged NSCLCs were significantly more likely to be found in younger patients and at an advanced stage; they showed cribriform features, extracellular mucus and psammoma bodies, whereas ROS1-discordant cases were found in older patients at a relatively early tumour-node-metastasis (TNM) stage and showed a lepidic growth pattern (all P < 0.001). Most ROS1-rearranged NSCLCs had no concurrent mutation, whereas 73% of discordant cases harboured genetic aberrations, including EGFR and ERBB2. Compared with general lung adenocarcinomas, ERBB-2 abnormality was disproportionately high in ROS1-discordant cases. Moreover, we optimised the scoring criteria for ROS1 IHC as 'H score > 150 and no concurrent mutations'; the specificity was then increased to 81.6%. CONCLUSIONS Compared with ROS1-rearranged cases, ROS1-discordant patients showed distinct clinical and morphological features and often harboured another oncogenic driver alteration. The use of optimised screening criteria will increase the specificity of ROS1 antibody.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaotong Chen
- Department of Pathology, The First Hospital of Yulin City, Guangxi, China
| | - Jing Zheng
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Mei Kong
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Bo Wang
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Ding
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
32
|
Torigoe H, Shien K, Takeda T, Yoshioka T, Namba K, Sato H, Suzawa K, Yamamoto H, Soh J, Sakaguchi M, Tomida S, Tsukuda K, Miyoshi S, Toyooka S. Therapeutic strategies for afatinib-resistant lung cancer harboring HER2 alterations. Cancer Sci 2018. [PMID: 29532558 PMCID: PMC5980184 DOI: 10.1111/cas.13571] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) plays an important role in the pathogenesis of various cancers. HER2 alterations have been suggested to be a therapeutic target in non-small-cell lung cancer (NSCLC), just as in breast and gastric cancers. We previously reported that the pan-HER inhibitor afatinib could be a useful therapeutic agent as HER2-targeted therapy for patients with NSCLC harboring HER2 alterations. However, acquired resistance to afatinib was observed in the clinical setting, similar to the case for other HER inhibitors. Thus, elucidation of the mechanisms underlying the development of acquired drug resistance and exploring means to overcome acquired drug resistance are important issues in the treatment of NSCLC. In this study, we experimentally established afatinib-resistant cell lines from NSCLC cell lines harboring HER2 alterations, and investigated the mechanisms underlying the acquisition of drug resistance. The established cell lines showed several unique afatinib-resistance mechanisms, including MET amplification, loss of HER2 amplification and gene expression, epithelial-to-mesenchymal transition (EMT) and acquisition of cancer stem cell (CSC)-like features. The afatinib-resistant cell lines showing MET amplification were sensitive to the combination of afatinib plus crizotinib (a MET inhibitor), both in vitro and in vivo. The resistant cell lines which showed EMT or had acquired CSC-like features remained sensitive to docetaxel, like the parental cells. These findings may provide clues to countering the resistance to afatinib in NSCLC patients with HER2 alterations.
Collapse
Affiliation(s)
- Hidejiro Torigoe
- Departments of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Clinical Genomic Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiko Shien
- Departments of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tatsuaki Takeda
- Clinical Pharmacy, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takahiro Yoshioka
- Clinical Genomic Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kei Namba
- Departments of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroki Sato
- Departments of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ken Suzawa
- Departments of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiromasa Yamamoto
- Departments of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Junichi Soh
- Departments of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masakiyo Sakaguchi
- Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shuta Tomida
- Biobank, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazunori Tsukuda
- Departments of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichiro Miyoshi
- Departments of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichi Toyooka
- Departments of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Clinical Genomic Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
33
|
Imperial R, Toor OM, Hussain A, Subramanian J, Masood A. Comprehensive pancancer genomic analysis reveals (RTK)-RAS-RAF-MEK as a key dysregulated pathway in cancer: Its clinical implications. Semin Cancer Biol 2017; 54:14-28. [PMID: 29175106 DOI: 10.1016/j.semcancer.2017.11.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/19/2017] [Indexed: 02/07/2023]
Abstract
Recent advances in Next Generation Sequencing (NGS) have provided remarkable insights into the genomic characteristics of human cancers that have spurred a revolution in the field of oncology. The mitogen-activated protein kinase pathway (MAPK) and its activating cell receptor, the receptor tyrosine kinases (RTKs), which together encompass the (RTK)-RAS-RAF-MEK-ERK axis, are central to oncogenesis. A pan-cancer genomics analysis presented in this review is made possible by large collaborative projects, including The Cancer Genome Atlas (TCGA), the International Cancer Genome Consortium (ICGC), and others. Landmark studies contributing to these projects have revealed alterations in cell signaling cascades that vary between cancer types and within tumors themselves. We review several of these studies in major tumor types to highlight recent advances in our understanding of the role of (RTK)-RAS-RAF alterations in cancer. Further studies are needed to increase the statistical power to detect clinically relevant low-frequency mutations, in addition to the known (RTK)-RAS-RAF pathway alterations, and to refine the resolution of the genomic landscape that defines these cancer mutations. The (RTK)-RAS-RAF-MEK-ERK mutation status, and their prognostic value, are also examined and correlated with clinical phenotypes. Treatments targeting various components of this pathway are ongoing, and are often effective initially in defined subgroups of patients. However, resistance to these agents can develop through adaptive mechanisms. With our steady increase in understanding the molecular biology of cancer, ongoing evaluation and monitoring through genomic analysis will continue to provide important information to the clinician in the context of treatment selection, response, resistance and outcomes.
Collapse
Affiliation(s)
- Robin Imperial
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Omer M Toor
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA; Division of Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA; Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA
| | - Arif Hussain
- Division of Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA; The Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Janakiraman Subramanian
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA; Division of Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA; Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA
| | - Ashiq Masood
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA; Division of Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA; Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA.
| |
Collapse
|
34
|
Abstract
The expanding spectrum of both established and candidate oncogenic driver mutations identified in non-small-cell lung cancer (NSCLC), coupled with the increasing number of clinically available signal transduction pathway inhibitors targeting these driver mutations, offers a tremendous opportunity to enhance patient outcomes. Despite these molecular advances, advanced-stage NSCLC remains largely incurable due to therapeutic resistance. In this Review, we discuss alterations in the targeted oncogene ('on-target' resistance) and in other downstream and parallel pathways ('off-target' resistance) leading to resistance to targeted therapies in NSCLC, and we provide an overview of the current understanding of the bidirectional interactions with the tumour microenvironment that promote therapeutic resistance. We highlight common mechanistic themes underpinning resistance to targeted therapies that are shared by NSCLC subtypes, including those with oncogenic alterations in epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), ROS1 proto-oncogene receptor tyrosine kinase (ROS1), serine/threonine-protein kinase b-raf (BRAF) and other less established oncoproteins. Finally, we discuss how understanding these themes can inform therapeutic strategies, including combination therapy approaches, and overcome the challenge of tumour heterogeneity.
Collapse
Affiliation(s)
- Julia Rotow
- Department of Medicine, Division of Hematology and Oncology, University of California San Francisco, 505 Parnassus Avenue, Box 1270, San Francisco, California 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, Box 0981, San Francisco, California 94143, USA
| | - Trever G Bivona
- Department of Medicine, Division of Hematology and Oncology, University of California San Francisco, 505 Parnassus Avenue, Box 1270, San Francisco, California 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, Box 0981, San Francisco, California 94143, USA
- Cellular and Molecular Pharmacology, University of California San Francisco, Box 2140, San Francisco, California 94158, USA
| |
Collapse
|
35
|
Lau KW, Seng C, Lim TKH, Tan DSW. Expanded molecular interrogation for potential actionable targets in non-squamous non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:372. [PMID: 29057232 PMCID: PMC5635263 DOI: 10.21037/atm.2017.08.42] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/03/2017] [Indexed: 12/15/2022]
Abstract
The advent of targeted therapies has established new standards of care for defined molecular subsets of non-small cell lung cancer (NSCLC). Not only has this led to significant changes in the routine clinical management of lung cancer e.g., multiplexed genomic testing, but it has provided important principles and benchmarks for determining "actionability". At present, the clinical paradigms are most evolved for EGFR mutations and ALK rearrangements, where multiple randomized phase III trials have determined optimal treatment strategies in both treatment naïve and resistant settings. However, this may not always be feasible with low prevalence alterations e.g., ROS1 and BRAF mutations. Another emerging observation is that not all targets are equally "actionable", necessitating a rigorous preclinical, clinical and translational framework to prosecute new targets and drug candidates. In this review, we will cover the role of targeted therapies for NSCLC harbouring BRAF, MET, HER2 and RET alterations, all of which have shown promise in non-squamous non-small cell lung cancer (ns-NSCLC). We further review some early epigenetic targets in NSCLC, an area of emerging interest. With increased molecular segmentation of lung cancer, we discuss the upcoming challenges in drug development and implementation of precision oncology approaches, especially in light of the complex and rapidly evolving therapeutic landscape.
Collapse
Affiliation(s)
- Kah Weng Lau
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
- Institute of Molecular and Cell Biology, ASTAR, Singapore
| | - Claudia Seng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Tony K H Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Cancer Therapeutics Research Laboratory, Singapore
- Genome Institute of Singapore, ASTAR, Singapore
| |
Collapse
|
36
|
Bruno TC, Ebner PJ, Moore BL, Squalls OG, Waugh KA, Eruslanov EB, Singhal S, Mitchell JD, Franklin WA, Merrick DT, McCarter MD, Palmer BE, Kern JA, Slansky JE. Antigen-Presenting Intratumoral B Cells Affect CD4 + TIL Phenotypes in Non-Small Cell Lung Cancer Patients. Cancer Immunol Res 2017; 5:898-907. [PMID: 28848053 DOI: 10.1158/2326-6066.cir-17-0075] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/10/2017] [Accepted: 08/21/2017] [Indexed: 12/25/2022]
Abstract
Effective immunotherapy options for patients with non-small cell lung cancer (NSCLC) are becoming increasingly available. The immunotherapy focus has been on tumor-infiltrating T cells (TILs); however, tumor-infiltrating B cells (TIL-Bs) have also been reported to correlate with NSCLC patient survival. The function of TIL-Bs in human cancer has been understudied, with little focus on their role as antigen-presenting cells and their influence on CD4+ TILs. Compared with other immune subsets detected in freshly isolated primary tumors from NSCLC patients, we observed increased numbers of intratumoral B cells relative to B cells from tumor-adjacent tissues. Furthermore, we demonstrated that TIL-Bs can efficiently present antigen to CD4+ TILs and alter the CD4+ TIL phenotype using an in vitro antigen-presentation assay. Specifically, we identified three CD4+ TIL responses to TIL-Bs, which we categorized as activated, antigen-associated, and nonresponsive. Within the activated and antigen-associated CD4+ TIL population, activated TIL-Bs (CD19+CD20+CD69+CD27+CD21+) were associated with an effector T-cell response (IFNγ+ CD4+ TILs). Alternatively, exhausted TIL-Bs (CD19+CD20+CD69+CD27-CD21-) were associated with a regulatory T-cell phenotype (FoxP3+ CD4+ TILs). Our results demonstrate a new role for TIL-Bs in NSCLC tumors in their interplay with CD4+ TILs in the tumor microenvironment, establishing them as a potential therapeutic target in NSCLC immunotherapy. Cancer Immunol Res; 5(10); 898-907. ©2017 AACR.
Collapse
Affiliation(s)
- Tullia C Bruno
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Peggy J Ebner
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Brandon L Moore
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Olivia G Squalls
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Katherine A Waugh
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Evgeniy B Eruslanov
- Division of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Division of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John D Mitchell
- Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Wilbur A Franklin
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniel T Merrick
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Martin D McCarter
- Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Brent E Palmer
- Division of Allergy and Clinical Immunology, University of Colorado School of Medicine, Aurora, Colorado
| | - Jeffrey A Kern
- Division of Oncology, National Jewish Health, Denver, Colorado
| | - Jill E Slansky
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
37
|
Pillai RN, Behera M, Berry LD, Rossi MR, Kris MG, Johnson BE, Bunn PA, Ramalingam SS, Khuri FR. HER2 mutations in lung adenocarcinomas: A report from the Lung Cancer Mutation Consortium. Cancer 2017; 123:4099-4105. [PMID: 28743157 DOI: 10.1002/cncr.30869] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/20/2017] [Accepted: 05/05/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2) mutations have been reported in lung adenocarcinomas. Herein, the authors describe the prevalence, clinical features, and outcomes associated with HER2 mutations in 1007 patients in the Lung Cancer Mutation Consortium (LCMC). METHODS Patients with advanced-stage lung adenocarcinomas were enrolled to the LCMC. Tumor specimens were assessed for diagnosis and adequacy; multiplexed genotyping was performed in Clinical Laboratory Improvement Amendments (CLIA)-certified laboratories to examine 10 oncogenic drivers. The LCMC database was queried for patients with HER2 mutations to access demographic data, treatment history, and vital status. An exploratory analysis was performed to evaluate the survival of patients with HER2 mutations who were treated with HER2-directed therapies. RESULTS A total of 920 patients were tested for HER2 mutations; 24 patients (3%) harbored exon 20 insertion mutations (95% confidence interval, 2%-4%). One patient had a concurrent mesenchymal-epithelial transition factor (MET) amplification. The median age of the patients was 62 years, with a slight predominance of females over males (14 females vs 10 males). The majority of the patients were never-smokers (71%) and presented with advanced disease at the time of diagnosis. The median survival for patients who received HER2-targeted therapies (12 patients) was 2.1 years compared with 1.4 years for those who did not (12 patients) (P = .48). Patients with HER2 mutations were found to have inferior survival compared with the rest of the LCMC cohort with other mutations: the median survival was 3.5 years in the LCMC population receiving targeted therapy and 2.4 years for patients not receiving targeted therapy. CONCLUSIONS HER2 mutations were detected in 3% of patients with lung adenocarcinoma in the LCMC. HER2-directed therapies should be investigated in this subgroup of patients. Cancer 2017;123:4099-4105. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Rathi N Pillai
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Madhusmita Behera
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Lynne D Berry
- Vanderbilt Center for Quantitative Sciences, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Mike R Rossi
- Division of Cancer Biology, Department of Radiation Oncology, Emory University, Atlanta, Georgia
| | - Mark G Kris
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bruce E Johnson
- Clinical Research, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Paul A Bunn
- Division of Medical Oncology, University of Colorado Cancer Center, Aurora, Colorado
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Fadlo R Khuri
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
38
|
Kim EK, Kim KA, Lee CY, Shim HS. The frequency and clinical impact of HER2 alterations in lung adenocarcinoma. PLoS One 2017; 12:e0171280. [PMID: 28146588 PMCID: PMC5287480 DOI: 10.1371/journal.pone.0171280] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/18/2017] [Indexed: 11/19/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2 or ErbB2) can be overexpressed, amplified and/or mutated in malignant tumors, and is a candidate for therapeutic targeting. However, molecular associations and clinical significances of these alterations were controversial in lung cancer. In this study, we investigated the frequency and clinicopathological significance of HER2 dysregulation in patients with lung adenocarcinoma. HER2 protein overexpression, gene amplification, and gene mutation were evaluated by immunohistochemistry (IHC), silver in situ hybridization, and direct sequencing, respectively. The H-scoring method and American Society of Clinical Oncology/College of American Pathologists breast cancer guidelines were used to interpret IHC results. Genetic analyses of EGFR and KRAS mutations, and of ALK and ROS1 rearrangements, were also performed. Of the 321 adenocarcinoma patients identified, HER2 overexpression (H-score ≥200) and gene amplification were found in 6 (1.9%) and 46 (14.3%), respectively. HER2 overexpression was correlated with papillary predominant histology; furthermore, it indicated poor overall survival and was an independent prognostic factor. HER2 amplification was associated with pleural invasion and showed a tendency towards shorter overall and disease-free survival. High-level gene amplification (HER2/CEP17 ratio ≥5 or copy number ≥10) was a poor prognostic factor for disease-free survival. HER2 mutations were detected in 6.7% (7 of 104) of driver oncogene-negative adenocarcinomas. Our study suggests that HER2 overexpression or amplification is a poor prognostic factor in lung adenocarcinoma, although the frequency of such events is low. Since molecular targeted agents are being tested in clinical trials, awareness of the specific HER2 status can influence the prognostic stratification and treatment of patients with molecularly defined subsets of lung adenocarcinoma.
Collapse
Affiliation(s)
- Eun Kyung Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung A. Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Chang Young Lee
- Department of Cardiovascular and Thoracic Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyo Sup Shim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
39
|
张 权, 张 树. [Research Progress of Targeted Therapy for Anaplastic Lymphoma Kinase and Other Rare Driver Genes in Advanced Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 20:66-72. [PMID: 28103976 PMCID: PMC5973289 DOI: 10.3779/j.issn.1009-3419.2017.01.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 11/11/2016] [Accepted: 11/12/2016] [Indexed: 11/05/2022]
Abstract
Targeted therapy was one of the major treatments in advanced non-small cell lung cancer (NSCLC) with positive driver genes. This area of research progresses day by day, with novel target discoveries, novel drug development, and use of novel combination treatments. Researchers have also undergone deep investigation about the molecular mechanisms underlying inherent or acquired resistance to these targeted therapies. This review aimed to summarize the advanced developments of targeted therapy for anaplastic lymphoma kinase (ALK) and other rare driver genes in NSCLC.
Collapse
Affiliation(s)
- 权 张
- />101149 北京,首都医科大学附属北京胸科医院肿瘤内科Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - 树才 张
- />101149 北京,首都医科大学附属北京胸科医院肿瘤内科Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| |
Collapse
|
40
|
Bansal P, Osman D, Gan GN, Simon GR, Boumber Y. Recent Advances in Targetable Therapeutics in Metastatic Non-Squamous NSCLC. Front Oncol 2016; 6:112. [PMID: 27200298 PMCID: PMC4854869 DOI: 10.3389/fonc.2016.00112] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 04/19/2016] [Indexed: 11/13/2022] Open
Abstract
Lung adenocarcinoma is the most common subtype of non-small cell lung cancer (NSCLC). With the discovery of epidermal growth factor receptor (EGFR) mutations, anaplastic lymphoma kinase (ALK) rearrangements, and effective targeted therapies, therapeutic options are expanding for patients with lung adenocarcinoma. Here, we review novel therapies in non-squamous NSCLC, which are directed against oncogenic targets, including EGFR, ALK, ROS1, BRAF, MET, human epidermal growth factor receptor 2 (HER2), vascular endothelial growth factor receptor 2 (VEGFR2), RET, and NTRK. With the rapidly evolving molecular testing and development of new targeted agents, our ability to further personalize therapy in non-squamous NSCLC is rapidly expanding.
Collapse
Affiliation(s)
- Pranshu Bansal
- Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA; Hematology/Oncology Fellowship Program, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Diaa Osman
- Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA; Hematology/Oncology Fellowship Program, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Gregory N Gan
- Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA; Section of Radiation Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - George R Simon
- Department of Thoracic and Head/Neck Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Yanis Boumber
- Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA; Cancer Genetics, Epigenetics, and Genomics Research Program, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| |
Collapse
|
41
|
Zhang K, Wang H. [Role of HER2 in NSCLC]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2016; 18:644-51. [PMID: 26483338 PMCID: PMC6000084 DOI: 10.3779/j.issn.1009-3419.2015.10.08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
过去几年中, 随着分子靶向药物的引入, 非小细胞肺癌(non-small cell lung cancer, NSCLC)的药物治疗策略发生了巨大变化, 向基于组织学和分子水平的治疗手段转变。表皮生长因子受体(epidermal growth factor receptor, EGFR)突变、Kirsten鼠肉瘤(Kirsten rat sarcoma, KRAS)癌基因突变、间变淋巴瘤激酶(anaplastic lymphoma kinase, ALK)重排等的发现, 影响着NSCLC治疗的发展。最近, 对人表皮生长因子受体2(human epidermal growth factor receptor 2, HER2)研究重燃兴趣, 这一基因改变与NSCLC对不同酪氨酸激酶抑制剂(tyrosine kinase inhibitors, TKIs)的敏感性相关, 其具有可能的预测作用, HER2扩增可能是EGFR突变肿瘤对EGFR-TKIs获得性耐药的机制之一。其次, HER2突变可能阐明一条新的靶向治疗NSCLC的策略。本文将对NSCLC中HER2异常调节发挥的作用做一简要介绍。
Collapse
Affiliation(s)
- Kun Zhang
- Department of Lung Oncology, Affiliated Hospital of The PLA Military Academy of Medical Sciences, Beijing 100071, China
| | - Hong Wang
- Department of Lung Oncology, Affiliated Hospital of The PLA Military Academy of Medical Sciences, Beijing 100071, China
| |
Collapse
|
42
|
Abstract
Lung cancer is the leading cause of cancer-related deaths in United States, accounting for more than one-fourth of the deaths annually. Although comparatively rare and relatively less studied, genetic abnormalities other than epidermal growth factor receptor (EGFR) mutations, anaplastic lymphoma kinase (ALK) rearrangements, and Kirsten rat sarcoma (KRAS) mutations account for significant proportion of the driver mutations identified thus far. The targeted agents against B-rapidly accelerated fibrosarcoma (BRAF) V600E mutation, MNNG-HOS transforming gene (MET) pathway, ROS1 rearrangement, rearranged during transfection (RET) rearrangement, and HER2 pathways offer promising therapeutic options. Recruiting patients with these rarer mutations to well-designed, large multicenter trials to further validate the use of targeted agents remains a challenge. The clinical data and ongoing trials with these agents are reviewed in this article.
Collapse
Affiliation(s)
- Nabin Khanal
- a Department of Internal Medicine , Creighton University Medical Center , Omaha , NE , USA
| | - Apar Kishor Ganti
- b Division of Oncology-Hematology, Department of Internal Medicine , VA-Nebraska Western Iowa Health Care System , Omaha , NE , USA.,c Division of Oncology-Hematology, Department of Internal Medicine , University of Nebraska Medical Center , Omaha , NE , USA
| |
Collapse
|
43
|
Lievense L, Aerts J, Hegmans J. Immune Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 893:59-90. [PMID: 26667339 DOI: 10.1007/978-3-319-24223-1_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lung cancer has long been considered an unsuitable target for immunotherapy due to its proposed immunoresistant properties. However, recent evidence has shown that anti-tumor immune responses can occur in lung cancer patients, paving the way for lung cancer as a novel target for immunotherapy. In order to take full advantage of the potential of immunotherapy, research is focusing on the presence and function of various immunological cell types in the tumor microenvironment. Immune cells which facilitate or inhibit antitumor responses have been identified and their prognostic value in lung cancer has been established. Knowledge regarding these pro- and anti-tumor immune cells and their mechanisms of action has facilitated the identification of numerous potential immunotherapeutic strategies and opportunities for intervention. A plethora of immunotherapeutic approaches is currently being developed and studied in lung cancer patients and phase 3 clinical trials are ongoing. Many different immunotherapies have shown promising clinical effects in patients with limited and advanced stage lung cancer, however, future years will have to tell whether immunotherapy will earn its place in the standard treatment of lung cancer.
Collapse
Affiliation(s)
- Lysanne Lievense
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Dr. Molewaterplein 50, Rotterdam, 3015 GD, The Netherlands
| | - Joachim Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Dr. Molewaterplein 50, Rotterdam, 3015 GD, The Netherlands
| | - Joost Hegmans
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Dr. Molewaterplein 50, Rotterdam, 3015 GD, The Netherlands.
| |
Collapse
|
44
|
Suzawa K, Toyooka S, Sakaguchi M, Morita M, Yamamoto H, Tomida S, Ohtsuka T, Watanabe M, Hashida S, Maki Y, Soh J, Asano H, Tsukuda K, Miyoshi S. Antitumor effect of afatinib, as a human epidermal growth factor receptor 2-targeted therapy, in lung cancers harboring HER2 oncogene alterations. Cancer Sci 2015; 107:45-52. [PMID: 26545934 PMCID: PMC4724821 DOI: 10.1111/cas.12845] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 10/21/2015] [Accepted: 10/31/2015] [Indexed: 01/26/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is a member of the HER family of proteins containing four receptor tyrosine kinases. It plays an important role in the pathogenesis of certain human cancers. In non-small-cell lung cancer (NSCLC), HER2 amplification or mutations have been reported. However, little is known about the benefit of HER2-targeted therapy for NSCLCs harboring HER2 alterations. In this study, we investigated the antitumor effect of afatinib, an irreversible epidermal growth factor receptor (EGFR)-HER2 dual inhibitor, in lung cancers harboring HER2 oncogene alterations, including novel HER2 mutations in the transmembrane domain, which we recently identified. Normal bronchial epithelial cells, BEAS-2B, ectopically overexpressing wild-type HER2 or mutants (A775insYVMA, G776VC, G776LC, P780insGSP, V659E, and G660D) showed constitutive autophosphorylation of HER2 and activation of downstream signaling. They were sensitive to afatinib, but insensitive to gefitinib. Furthermore, we examined the antitumor activity of afatinib and gefitinib in several NSCLC cell lines, and investigated the association between their genetic alterations and sensitivity to afatinib treatment. In HER2-altered NSCLC cells (H2170, Calu-3, and H1781), afatinib downregulated the phosphorylation of HER2 and EGFR as well as their downstream signaling, and induced an antiproliferative effect through G1 arrest and apoptotic cell death. In contrast, HER2- or EGFR-non-dependent NSCLC cells were insensitive to afatinib. In addition, these effects were confirmed in vivo by using a xenograft mouse model of HER2-altered lung cancer cells. Our results suggest that afatinib is a therapeutic option as a HER2-targeted therapy for NSCLC harboring HER2 amplification or mutations.
Collapse
Affiliation(s)
- Ken Suzawa
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichi Toyooka
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Clinical Genomic Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Biobank, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masakiyo Sakaguchi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mizuki Morita
- Department of Biobank, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Biorepository Research and Networking, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiromasa Yamamoto
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shuta Tomida
- Department of Biobank, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tomoaki Ohtsuka
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mototsugu Watanabe
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinsuke Hashida
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Clinical Genomic Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuho Maki
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Junichi Soh
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Biobank, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroaki Asano
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazunori Tsukuda
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichiro Miyoshi
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
45
|
Mazières J, Barlesi F, Filleron T, Besse B, Monnet I, Beau-Faller M, Peters S, Dansin E, Früh M, Pless M, Rosell R, Wislez M, Fournel P, Westeel V, Cappuzzo F, Cortot A, Moro-Sibilot D, Milia J, Gautschi O. Lung cancer patients with HER2 mutations treated with chemotherapy and HER2-targeted drugs: results from the European EUHER2 cohort. Ann Oncol 2015; 27:281-6. [PMID: 26598547 DOI: 10.1093/annonc/mdv573] [Citation(s) in RCA: 238] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/07/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND HER2 mutations have been identified as oncogenic drivers in lung cancers and are found in 1-2% of lung adenocarcinomas. There is, to date, no standard of care for these patients. We thus aim to study the therapeutic outcomes of patients harboring HER2 mutations and establish the efficacy of various drug regimens. PATIENTS AND METHODS This retrospective cohort study in European centers assessed patients with advanced non-small-cell lung cancer (NSCLC), a known HER2 exon-20 insertion, treated with chemotherapy and/or HER2-targeted drugs. RESULTS We identified 101 eligible patients from 38 centers: median age 61 years (range: 30-87), 62.4% women, 60.4% never-smokers. All tumors were adenocarcinomas. Concomitant EGFR mutations, ALK translocations, and ROS translocations were observed in 5, 1, and 1 patients, respectively. The median number of treatment lines was 3 (range: 1-11). The median overall survival was 24 months. Overall response rate (ORR) and the median progression-free survival (PFS) with conventional chemotherapy (excluding targeted therapies) were 43.5% and 6 months in first-line (n = 93), and 10% and 4.3 months in second-line (n = 52) therapies. Sixty-five patients received HER2-targeted therapies: trastuzumab = 57, neratinib = 14, afatinib = 9, lapatinib = 5, T-DM1 = 1. ORR was 50.9% and PFS was 4.8 months with trastuzumab or T-DM1. CONCLUSION This series shows the chemosensitivity of HER2-driven NSCLC, and the potential interest of HER2-targeted agents. Our results should help to define the best therapeutic strategy for these patients and to orient future clinical trials.
Collapse
Affiliation(s)
- J Mazières
- Thoracic Oncology Unit, Larrey Hospital, Centre Hospitalier Universitaire de Toulouse, Toulouse University III (Paul Sabatier), Toulouse
| | - F Barlesi
- Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille
| | - T Filleron
- Clinical Research Unit, Biostatistics, Cancer University Institute of Toulouse Oncopole, Toulouse
| | - B Besse
- Thoracic Oncology Section, Gustave Roussy Cancer Campus, Villejuif
| | - I Monnet
- Pulmonology Department, Centre hospitalier intercommunal, Créteil
| | - M Beau-Faller
- Chest Department, Strasbourg University Hospital, Strasbourg, France
| | - S Peters
- Medical Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - E Dansin
- Medical Oncology Department, Centre Oscar Lambret, Lille, France
| | - M Früh
- Department of Oncology and Hematology, Cantonal Hospital of St Gallen, St Gallen
| | - M Pless
- Department of Medical Oncology, Kantonsspital Winterthur, Winterthur, Switzerland
| | - R Rosell
- Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Barcelona, Spain
| | - M Wislez
- Pulmonary Medicine Unit, AP-HP, Hôpital Tenon, Paris
| | - P Fournel
- Medical Oncology Department, Lucien Neuwirth Cancer Institute, Saint Priest en Jarez
| | - V Westeel
- Pulmonology Department, Centre Hospitalier Régional Universitaire, Hôpital Jean Minjoz, Besançon, France
| | - F Cappuzzo
- Medical Oncology Department, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy
| | - A Cortot
- Pulmonology Department, Centre Hospitalier Universitaire, Lille
| | - D Moro-Sibilot
- Department of Thoracic Oncology, Centre Hospitalier Universitaire, Grenoble, France
| | - J Milia
- Thoracic Oncology Unit, Larrey Hospital, Centre Hospitalier Universitaire de Toulouse, Toulouse University III (Paul Sabatier), Toulouse
| | - O Gautschi
- Medical Oncology, Cantonal Hospital Luzern, Luzern, Switzerland
| |
Collapse
|
46
|
Abstract
The therapeutic landscape of non-small-cell lung cancer (NSCLC) has dramatically changed in the last few years with the introduction of molecularly targeted agents, leading to unprecedented results in lung tumors with a paradigmatic shift from a "one size fits all" approach to an histologic and molecular-based approach. The discovery of epidermal growth factor receptor (EGFR) mutations in NSCLC in 2004 and the marked response to the EGFR tyrosine kinase inhibitor gefitinib, in a small subset of patients harboring these genetic abnormalities, stimulated the study of other kinase mutants involvement in NSCLC. The incredible story of ALK rearranged tumors, with the rapid Food and Drug Administration approval of Crizotinib after only 4 years from the discovery of EML4-ALK translocation in NSCLC, has profoundly influenced the concept of drug development in NSCLC, paving the way to a novel series of molecularly selected studies with specific inhibitors. The identification of these oncogenic drivers has dramatically changed the genetic landscape of NSCLC moving away from the old concept of a large indistinct histological entity to a combination of rare clinically relevant molecular subsets. Recently, a renewed interest has been emerging on the human epidermal growth factor-2 (HER2) pathway. Genetic aberrations of this signaling pathway have been reported over time to be associated in NSCLC with different sensitivity to the EGFR tyrosine kinase inhibitors, to have a possible prognostic role and more recently HER2 amplification has been emerged as a possible mechanism in EGFR-mutated tumors of acquired resistance to the EGFR tyrosine kinase inhibitors. In addition, dysregulation of the HER2 pathway, in particular HER2 mutations (mostly, in-frame exon 20 insertions), may represent a possible novel therapeutic target in NSCLC, paving the way for a new generation of targeted agents in NSCLC. Since anecdotal case reports of clinical activity of anti-HER2 agents in NSCLC patients with HER2 mutations, several targeted agents have been evaluated in HER2-mutated patients, generating a growing interest upon this oncogenic driver, leading to the design of molecularly selected trials with anti-HER2 compounds and the rediscover of hastily thrown out drugs, such as neratinib. The aim of this article is to provide an overview of the role of HER2 dysregulation in NSCLCs, trying to throw a light not only on the strengths but also the weaknesses of the studies conducted so far. It is a long way to the clinical implementation of these biomarkers and probably the increasing use of next generation sequencing techniques, the creation of large multi-institutional molecular testing platforms and the design of rationally based trials can get closer personalized medicine in NSCLC.
Collapse
|
47
|
Teixidó C, Karachaliou N, González-Cao M, Morales-Espinosa D, Rosell R. Assays for predicting and monitoring responses to lung cancer immunotherapy. Cancer Biol Med 2015; 12:87-95. [PMID: 26175924 PMCID: PMC4493376 DOI: 10.7497/j.issn.2095-3941.2015.0019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 04/30/2015] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy has become a key strategy for cancer treatment, and two immune checkpoints, namely, programmed cell death 1 (PD-1) and its ligand (PD-L1), have recently emerged as important targets. The interaction blockade of PD-1 and PD-L1 demonstrated promising activity and antitumor efficacy in early phase clinical trials for advanced solid tumors such as non-small cell lung cancer (NSCLC). Many cell types in multiple tissues express PD-L1 as well as several tumor types, thereby suggesting that the ligand may play important roles in inhibiting immune responses throughout the body. Therefore, PD-L1 is a critical immunomodulating component within the lung microenvironment, but the correlation between PD-L1 expression and prognosis is controversial. More evidence is required to support the use of PD-L1 as a potential predictive biomarker. Clinical trials have measured PD-L1 in tumor tissues by immunohistochemistry (IHC) with different antibodies, but the assessment of PD-L1 is not yet standardized. Some commercial antibodies lack specificity and their reproducibility has not been fully evaluated. Further studies are required to clarify the optimal IHC assay as well as to predict and monitor the immune responses of the PD-1/PD-L1 pathway.
Collapse
Affiliation(s)
- Cristina Teixidó
- 1 Pangaea Biotech, Quirón Dexeus University Hospital, Barcelona 08028, Spain ; 2 Dr. Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona 08028, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain
| | - Niki Karachaliou
- 1 Pangaea Biotech, Quirón Dexeus University Hospital, Barcelona 08028, Spain ; 2 Dr. Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona 08028, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain
| | - Maria González-Cao
- 1 Pangaea Biotech, Quirón Dexeus University Hospital, Barcelona 08028, Spain ; 2 Dr. Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona 08028, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain
| | - Daniela Morales-Espinosa
- 1 Pangaea Biotech, Quirón Dexeus University Hospital, Barcelona 08028, Spain ; 2 Dr. Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona 08028, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain
| | - Rafael Rosell
- 1 Pangaea Biotech, Quirón Dexeus University Hospital, Barcelona 08028, Spain ; 2 Dr. Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona 08028, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain
| |
Collapse
|
48
|
McCoach CE, Doebele RC. The minority report: targeting the rare oncogenes in NSCLC. Curr Treat Options Oncol 2015; 15:644-57. [PMID: 25228144 DOI: 10.1007/s11864-014-0310-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OPINION STATEMENT Lung cancer is still responsible for the highest number of cancer deaths worldwide. Despite this fact, significant progress has been made in the treatment of non-small cell lung cancer (NSCLC). Specifically, efforts to identify and treat genetic alterations (gene mutations, gene fusions, gene amplification events, etc.) that result in oncogenic drivers are now standard of care (EGFR and ALK) or an intense area of research. The most prevalent oncogenic drivers have likely already been identified; thus, there is now a focus on subgroups of tumors with less common genetic alterations. Interestingly, as we explore these less common mutations, we are discovering that many occur across other tumor types (i.e., non-lung cancer), further justifying their study. Furthermore, many studies have demonstrated that by searching broadly for multiple genetic alterations in large subsets of patients they are able to identify potentially targetable alterations in the majority of patients. Although individually, the rare oncogenic drivers subgroups may seem to occur too infrequently to justify their exploration, the fact that the majority of patients with NSCLC harbor a potentially actionable driver mutation within their tumors and the fact that different types of cancers often have the same oncogenic driver justifies this approach.
Collapse
Affiliation(s)
- Caroline E McCoach
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, 12801 E. 17th Avenue, Aurora, CO, 80045, USA
| | | |
Collapse
|
49
|
Hall PE, Spicer J, Popat S. Rationale for targeting the ErbB family of receptors in patients with advanced squamous cell carcinoma of the lung. Future Oncol 2015; 11:2175-91. [PMID: 26039665 DOI: 10.2217/fon.15.110] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Squamous cell carcinoma (SCC) of the lung represents around 30% of all non-small-cell lung cancers. Treatment options for nonsquamous histology have increased in recent years following the development of pemetrexed chemotherapy and the identification of activating EGFR mutations and ALK rearrangements as targets for effective noncytotoxic agents. By contrast, until recently the development of new therapies for SCC has lagged behind. However, the identification of important genetic events driving SCC, including a greater understanding of the role of the ErbB receptor family in SCC pathogenesis, as well as recent immunotherapy advances, have led to new treatment options for SCC.
Collapse
Affiliation(s)
- Peter E Hall
- Department of Medical Oncology, Guy's & St Thomas' NHS Trust, London, UK
| | - James Spicer
- Department of Medical Oncology, Guy's & St Thomas' NHS Trust, London, UK.,Division of Cancer Studies, King's College London, London, UK
| | | |
Collapse
|
50
|
Califano R, Abidin A, Tariq NUA, Economopoulou P, Metro G, Mountzios G. Beyond EGFR and ALK inhibition: Unravelling and exploiting novel genetic alterations in advanced non small-cell lung cancer. Cancer Treat Rev 2015; 41:401-11. [DOI: 10.1016/j.ctrv.2015.03.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 03/03/2015] [Accepted: 03/22/2015] [Indexed: 02/07/2023]
|