CC BY 4.0 · Glob Med Genet 2024; 11(01): 086-099
DOI: 10.1055/s-0044-1781457
Review Article

Expert Consensus on the Diagnosis and Treatment of NRG1/2 Gene Fusion Solid Tumors

Chunwei Xu*
1   Department of Scientific Research, Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou Zhejiang, People's Republic of China
2   Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Qian Wang*
3   Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing Jiangsu, People's Republic of China
,
Dong Wang*
2   Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Wenxian Wang*
4   Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, People's Republic of China
,
Wenfeng Fang*
5   Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou Guangdong, People's Republic of China
,
Ziming Li
6   Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
,
Aijun Liu
7   Senior Department of Pathology, the 7th Medical Center of PLA General Hospital, Beijing, People's Republic of China
,
Jinpu Yu
8   Department of Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
,
Wenzhao Zhong
9   Department of Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangzhou Guangdong, People's Republic of China
,
Zhijie Wang
10   State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
,
Yongchang Zhang
11   Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha Hunan, People's Republic of China
,
Jingjing Liu
12   Department of Thoracic Cancer, Jilin Cancer Hospital, Jilin Changchun, People's Republic of China
,
Shirong Zhang
13   Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou Zhejiang, People's Republic of China
,
Xiuyu Cai
14   Department of VIP Inpatient, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, People's Republic of China
,
Anwen Liu
15   Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang Jiangxi, People's Republic of China
,
Wen Li
16   Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou Zhejiang, People's Republic of China
,
Ping Zhan
2   Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Hongbing Liu
2   Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Tangfeng Lv
2   Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Liyun Miao
17   Department of Respiratory Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Lingfeng Min
18   Department of Respiratory Medicine, Clinical Medical School of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou Jiangsu, People's Republic of China
,
Yu Chen
19   Department of Medical Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou Fujian, People's Republic of China
,
Jingping Yuan
20   Department of Pathology, Renmin Hospital of Wuhan University, Wuhan Hubei, People's Republic of China
,
Feng Wang
21   Department of Internal Medicine, Cancer Center of PLA, Qinhuai Medical Area, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, People's Republic of China
,
Zhansheng Jiang
22   Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
,
Gen Lin
19   Department of Medical Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou Fujian, People's Republic of China
,
Long Huang
15   Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang Jiangxi, People's Republic of China
,
Xingxiang Pu
23   Department of Medical Oncology, Lung Cancer and Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha Hunan, People's Republic of China
,
Rongbo Lin
19   Department of Medical Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou Fujian, People's Republic of China
,
Weifeng Liu
24   Department of Orthopaedic Oncology Surgery, Beijing Ji Shui Tan Hospital, Peking University, Beijing, People's Republic of China
,
Chuangzhou Rao
25   Department of Radiotherapy and Chemotherapy, Hwamei Hospital, University of Chinese Academy of Sciences, Ningbo Zhejiang, People's Republic of China
,
Dongqing Lv
26   Department of Pulmonary Medicine, Taizhou Hospital of Wenzhou Medical University, Taizhou Zhejiang, People's Republic of China
,
Zongyang Yu
27   Department of Respiratory Medicine, the 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital), Fujian Medical University, Fuzhou Fujian, People's Republic of China
,
Xiaoyan Li
28   Department of Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
,
Chuanhao Tang
29   Department of Medical Oncology, Peking University International Hospital, Beijing, People's Republic of China
,
Chengzhi Zhou
30   Department of State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University (The First Affiliated Hospital of Guangzhou Medical University), Guangzhou Guangdong, People's Republic of China
,
Junping Zhang
31   Department of Thoracic Oncology, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Taiyuan Shanxi, People's Republic of China
,
Junli Xue
32   Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
,
Hui Guo
33   Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi, People's Republic of China
,
Qian Chu
34   Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, People's Republic of China
,
Rui Meng
35   Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, People's Republic of China
,
Jingxun Wu
36   Department of Medical Oncology, the First Affiliated Hospital of Medicine, Xiamen University, Xiamen Fujian, People's Republic of China
,
Rui Zhang
37   Department of Medical Oncology, Cancer Hospital of China Medical University, Shenyang Liaoning, People's Republic of China
,
Jin Zhou
38   Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology, Chengdu Sichuan, People's Republic of China
,
Zhengfei Zhu
39   Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
,
Yongheng Li
40   Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
,
Hong Qiu
34   Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, People's Republic of China
,
Fan Xia
39   Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
,
Yuanyuan Lu
41   Department of State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an Shaanxi, People's Republic of China
,
Xiaofeng Chen
42   Department of Oncology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing Jiangsu, People's Republic of China
,
Rui Ge
43   Department of General Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
,
Enyong Dai
44   Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun Jilin, People's Republic of China
,
Yu Han
45   Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin Heilongjiang, People's Republic of China
,
Weiwei Pan
46   Department of Cell Biology, College of Medicine, Jiaxing University, Jiaxing Zhejiang, People's Republic of China
,
Fei Pang
47   Department of Medical, Shanghai OrigiMed Co., Ltd., Shanghai, People's Republic of China
,
Qingqing He
47   Department of Medical, Shanghai OrigiMed Co., Ltd., Shanghai, People's Republic of China
,
Jintao Huang
47   Department of Medical, Shanghai OrigiMed Co., Ltd., Shanghai, People's Republic of China
,
Kai Wang
47   Department of Medical, Shanghai OrigiMed Co., Ltd., Shanghai, People's Republic of China
,
Fan Wu
48   Department of Medical, Stone Pharmaceuticals (Suzhou) Co., Ltd., Shanghai, People's Republic of China
,
Bingwei Xu
49   Department of Biotherapy, Cancer Institute, First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
,
Liping Wang
50   Department of Oncology, Baotou Cancer Hospital, Baotou Inner Mongolia, People's Republic of China
,
Youcai Zhu
51   Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital, The Third Affiliated Hospital of Jiaxing University, Jiaxing Zhejiang, People's Republic of China
,
Li Lin
29   Department of Medical Oncology, Peking University International Hospital, Beijing, People's Republic of China
,
Yanru Xie
52   Department of Oncology, Lishui Municipal Central Hospital, Lishui Zhejiang, People's Republic of China
,
Xinqing Lin
30   Department of State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University (The First Affiliated Hospital of Guangzhou Medical University), Guangzhou Guangdong, People's Republic of China
,
Jing Cai
15   Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang Jiangxi, People's Republic of China
,
Ling Xu
53   Department of Interventional Pulmonary Diseases, Anhui Chest Hospital, Hefei Anhui, People's Republic of China
,
Jisheng Li
54   Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinnan Shangdong, People's Republic of China
,
Xiaodong Jiao
55   Department of Medical Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, People's Republic of China
,
Kainan Li
56   Department of Oncology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan Shandong, People's Republic of China
,
Jia Wei
57   Department of the Comprehensive Cancer Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Huijing Feng
31   Department of Thoracic Oncology, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Taiyuan Shanxi, People's Republic of China
,
Lin Wang
58   Department of Pathology, Shanxi Academy of Medical Sciences, Shanxi Bethune Hospital, Taiyuan Shanxi, People's Republic of China
,
Yingying Du
59   Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei Anhui, People's Republic of China
,
Wang Yao
60   Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou Guangdong, People's Republic of China
,
Xuefei Shi
61   Department of Respiratory Medicine, Huzhou Hospital, Zhejiang University School of Medicine, Huzhou Zhejiang, People's Republic of China
,
Xiaomin Niu
6   Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
,
Dongmei Yuan
2   Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Yanwen Yao
2   Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Jianhui Huang
52   Department of Oncology, Lishui Municipal Central Hospital, Lishui Zhejiang, People's Republic of China
,
Yue Feng
62   Department of Gynecologic Radiation Oncology, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, People's Republic of China
,
Yinbin Zhang
63   Department of Oncology, the Second Affiliated Hospital of Medical College, Xi′an Jiaotong University, Xi'an Shaanxi, People's Republic of China
,
Pingli Sun
64   Department of Pathology, The Second Hospital of Jilin University, Changchun Jilin, People's Republic of China
,
Hong Wang
65   Senior Department of Oncology, The 5th Medical Center of PLA General Hospital, Beijing, People's Republic of China
,
Mingxiang Ye
2   Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Zhaofeng Wang
2   Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Yue Hao
4   Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, People's Republic of China
,
Zhen Wang
66   Department of Radiation Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, People's Republic of China
,
Bin Wan
67   Department of Respiratory Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing Jiangsu, People's Republic of China
,
Donglai Lv
68   Department of Clinical Oncology, The 901 Hospital of Joint Logistics Support Force of People Liberation Army, Hefei Anhui, People's Republic of China
,
Shengjie Yang
69   Department of Thoracic Surgery, Chuxiong Yi Autonomous Prefecture People's Hospital, Chuxiong, People's Republic of China
,
Jin Kang
9   Department of Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangzhou Guangdong, People's Republic of China
,
Jiatao Zhang
9   Department of Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangzhou Guangdong, People's Republic of China
,
Chao Zhang
9   Department of Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangzhou Guangdong, People's Republic of China
,
Juanjuan Ou
70   Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
,
Lin Shi
71   Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
,
Yina Wang
72   Department of Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou Zhejiang, People's Republic of China
,
Bihui Li
73   Department of Oncology, The Second Affiliated Hospital of Guilin Medical University, Guilin Guangxi, People's Republic of China
,
Zhang Zhang
74   Department of International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou, Guangdong, People's Republic of China
,
Zhongwu Li
75   Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
,
Zhefeng Liu
65   Senior Department of Oncology, The 5th Medical Center of PLA General Hospital, Beijing, People's Republic of China
,
Nong Yang
11   Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha Hunan, People's Republic of China
,
Lin Wu
23   Department of Medical Oncology, Lung Cancer and Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha Hunan, People's Republic of China
,
Huijuan Wang
76   Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou Henan, People's Republic of China
,
Gu Jin
77   Department of Bone and Soft-tissue Surgery, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou Zhejiang, People's Republic of China
,
Guansong Wang
78   Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
,
Jiandong Wang
79   Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, People's Republic of China
,
Meiyu Fang
3   Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing Jiangsu, People's Republic of China
,
Yong Fang
80   Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou Zhejiang, People's Republic of China
,
Yuan Li
81   Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
,
Xiaojia Wang
4   Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, People's Republic of China
,
Yiping Zhang
4   Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, People's Republic of China
,
Xixu Zhu
66   Department of Radiation Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, People's Republic of China
,
Yi Shen
82   Department of Thoracic Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Shenglin Ma
83   Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou Zhejiang, People's Republic of China
,
Biyun Wang
84   Department of Breast Cancer and Urological Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
,
Lu Si
85   Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
,
Yong Song
2   Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing Jiangsu, People's Republic of China
,
Yuanzhi Lu
86   Department of Clinical Pathology, the First Affiliated Hospital of Jinan University, Guangzhou Guangdong, People's Republic of China
,
Jing Chen
35   Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, People's Republic of China
,
Zhengbo Song
4   Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, People's Republic of China
› Author Affiliations
Funding This work was supported by the Natural Science Foundation of China (grant numbers 82002456), China Postdoctoral Science Foundation (grant numbers 2022M723207), the Medical Scientific Research Foundation of Zhejiang Province of China (grant numbers 2023KY666), Zhejiang Traditional Chinese Medicine Science Fund Project (grant numbers 2024ZL372), Qiantang Cross Fund Project (grant numbers 2023-16), National Natural Science Foundation of China of Zhejiang Cancer Hospital Cultivation Project (grant numbers PY2023006), the Medical Scientific Research Foundation of Zhejiang Province of China (grant numbers 2024KY812), and The Natural Science Foundation of Zhejiang Province (grant numbers Q24H160110).
 

Abstract

The fusion genes NRG1 and NRG2, members of the epidermal growth factor (EGF) receptor family, have emerged as key drivers in cancer. Upon fusion, NRG1 retains its EGF-like active domain, binds to the ERBB ligand family, and triggers intracellular signaling cascades, promoting uncontrolled cell proliferation. The incidence of NRG1 gene fusion varies across cancer types, with lung cancer being the most prevalent at 0.19 to 0.27%. CD74 and SLC3A2 are the most frequently observed fusion partners. RNA-based next-generation sequencing is the primary method for detecting NRG1 and NRG2 gene fusions, whereas pERBB3 immunohistochemistry can serve as a rapid prescreening tool for identifying NRG1-positive patients. Currently, there are no approved targeted drugs for NRG1 and NRG2. Common treatment approaches involve pan-ERBB inhibitors, small molecule inhibitors targeting ERBB2 or ERBB3, and monoclonal antibodies. Given the current landscape of NRG1 and NRG2 in solid tumors, a consensus among diagnostic and treatment experts is proposed, and clinical trials hold promise for benefiting more patients with NRG1 and NRG2 gene fusion solid tumors.


#

Introduction

Gene fusion caused by chromosomal rearrangement is a common event in solid tumors, driving tumorigenesis. The identification and targeting of fusion genes have been significant breakthroughs in medicine. Chromosomal rearrangements of receptor tyrosine kinases (RTKs) can generate oncogenic fusion protein kinases. Several tyrosine kinase inhibitors (TKIs) have been approved for treating solid malignancies with RTK fusions.[1] The epidermal growth factor (EGF) receptor family belongs to the type I RTK family. NRG1 and NRG2 genes encode neuroregulin 1 and 2 proteins, respectively, which are part of the EGF ligand family. NRG1 gene fusion activates and retains the EGF-like domain of the NRG1 protein, continuously binding to ERBB receptor family members (ERBB2 and ERBB4). This initiates intracellular signaling cascades, leading to sustained cell proliferation and tumorigenesis.[2]

Although NRG1 gene fusion in solid tumors is rare (0.2%), patients with NRG1 fusion tumors often have a poor response to standard treatments. Disrupting NRG1 binding to ERBB3 or impacting ERBB2/ERBB3 heterodimerization can reduce the volume of NRG1 fusion tumors in various solid tumors.[3] NRG1 is an emerging oncogenic driver and a potential therapeutic target, but no approved targeted drugs are available for NRG1 fusion tumors. NRG2 fusion has also been found in lung adenocarcinoma patients, but further understanding of its biological functions is needed.[4] [5]

This article summarizes the biological behaviors of NRG1 and NRG2 fusion-related proteins and introduces molecular characteristic data of NRG1 gene fusion in solid tumors from the largest-scale database. It proposes a screening strategy for NRG1/2 gene fusion solid tumors based on existing domestic resources. Ongoing clinical trials targeting NRG1 fusion solid tumors are also summarized, along with proposed treatment consensus.


#

The Biological Basis of the NRG1/2 Gene

The Gene Structures and Biological Functions of the NRG1/2 Gene

RTKs are essential in drug development, with the ERBB family, including ERBB1 (EGFR), ERBB2 (HER2), ERBB3 (HER3), and ERBB4 (HER4), being transmembrane RTKs known as the EGF receptor family. The tyrosine kinase ligand family, which includes the neuregulin family (NRGs), consists of six protein isoforms: NRG1, NRG2, NRG3, NRG4, NRG5 (tomoregulin), and NRG6 (neuroglycan C). These ligands all contain an extracellular EGF-like domain that activates the ERBB RTK. They are crucial for the development of the nervous and cardiovascular systems.[6] [7]

NRG1

The NRG1 gene, also known as Neuregulin 1, Heregulin, Neu differentiation factor, Glial growth factor, and Acetylcholine receptor-inducing activity, is located at 8p21.[8] [9] [10] [11] [12] NRG1 interacts with ERBB3 and ERBB4 through its EGF-like domain, tissue specificity, and immunoglobulin-like domain.[13] NRG1 has multiple isoforms and structural differences, with six protein subtypes (I–VI) and at least 31 gene subtypes. The NRG1 protein consists of the EGF-like domain, the N-terminal sequence (type I, II, or III), and the C-terminal sequence (transmembrane or not). Type I and II NRGs are also referred to as “Ig-NRGs,” whereas type III NRGs are known as “CRD-NRGs.” The fusion-involved subtype of NRG1 belongs to type III and has a higher affinity for receptor binding than the α-type. This difference in binding affinity contributes to the oncogenic properties of NRG1 IIIβ compared with NRG1 IIIα. NRG1 is initially produced as a membrane-anchored precursor, and proteolysis releases the EGF-like domain, activating ERBB3 and ERBB4. The interaction between NRG1 and ERBB3 can lead to heterodimerization, particularly with ERBB2, facilitating downstream signaling pathways such as PI3K/AKT and MAPK. NRG1 can also interact with ERBB4, forming homodimers or heterodimers with ERBB2/ERBB3, further activating multiple pathways[14] [15] ([Fig. 1A]).

Zoom Image
Fig. 1 NRG1 and NRG2 structures. (A) NRG1 possesses I, II, and III subtype structures patterns. The coding sequences of the same isoform vary due to diverse transcription start sites and alternative splicing of NRG1 gene promoters. It is worth noting that the EGF-like domain alone has the capability to efficiently activate homologous ERBB receptor tyrosine kinases. N and C marked in red represent the N-terminal and C-terminal of NRG1 protein, respectively. To obtain further information, please refer to the relevant literature.[2] [38] (B) NRG2 structure. CRD, cysteine-rich domain; CTc, cytoplasmic tail domain C terminal of the EGF-like domain; TMc, transmembrane domain C terminal of the EGF-like domain; TMn, transmembrane domain N-terminal of the EGF-like domain.

#

NRG2

The NRG2 gene, also known as Divergent of neuregulin 1, Neural and thymus derived activator for ErbB kinases, and Neuregulin 2, is located at 5q13.2.[16] [17] [18] NRG2 has two isoforms, α and β, due to different splicing sites. Research has shown that NRG2β is a high-affinity ligand for ERBB4, strongly stimulating ERBB4 tyrosine phosphorylation. On the other hand, the splicing isoform NRG2α is a low-affinity ligand for ERBB4 and does not strongly stimulate ERBB4 phosphorylation[19] ([Fig. 1B]).


#
#

Fusion and Carcinogenic Mechanism of NRG1/2

The activation or overexpression of NRGs has been shown to regulate tumor cell growth, invasion, and angiogenesis. These genes are associated with various types of tumors including breast cancer, ovarian cancer, endometrial cancer, colorectal cancer, gastric cancer, lung cancer, thyroid cancer, glioma, medulloblastoma, melanoma, and head and neck squamous cell carcinoma.[8] [20] [21] In solid tumors, gene fusion is a significant driver mutation. Specifically, NRG1 gene fusion is considered a potential targetable oncogenic driver. The oncogenicity of NRG1 and NRG2 gene fusions relies on maintaining an intact EGF-like domain without frameshift mutations.[2] Knockout mouse models with disrupted EGF-like domain (neuregulinδEGF-LacZ) have demonstrated that all NRG1 subtypes lose their function, leading to embryonic death due to cardiac and nervous system malformations.[22]

The discovery of NRG1 fusion dates back to 1997 in the breast cancer cell line MDA-MB-175, where it was identified as a tumor-specific DOC4-NRG1 transcript that promotes tumor cell proliferation.[23] In lung cancer, NRG1 gene fusion results in the overexpression of the EGF-like domain of NRG1 on the cell surface. This enhances its binding ability with ERBB3, promoting heterodimerization of ERBB2/ERBB3 and subsequently activating downstream PI3K/AKT and MAPK signaling pathways.[24] Studies using CD74-NRG1 transgenic mouse models have shown that the proliferation of CD74-NRG1 cells is carcinogenic and accompanied by increased protein transcription levels of ERBB2 and ERBB3, indicating that NRG1 gene fusion drives tumor development.[25] NRG1 gene fusion is the first potential therapeutic oncogenic driver mutation specifically associated with a subtype of lung adenocarcinoma and is predominantly found in nonsmoking patients, in contrast to the tobacco-associated KRAS gene mutation.[24] In a transcriptome sequencing study of 25 never-smoking lung adenocarcinoma patients, one case of CD74-NRG1 gene fusion was identified in a patient with invasive mucinous subtype. Mechanistically, CD74-NRG1 gene fusion leads to extracellular expression of the EGF-like domain of NRG1 III-β3, providing a ligand for the ERBB2–ERBB3 receptor complex. Consequently, ERBB2 and ERBB3 are highly expressed in index cases, and phosphorylated ERBB3 is specifically expressed in fusion tumors (p < 0.0001). In lung cancer cell lines expressing ERBB2 and ERBB3, ectopic expression of CD74-NRG1 activates the ERBB3 and PI3K-AKT pathways, resulting in increased colony formation in soft agar.[26]

Breakpoints on the NRG1 chromosome were discovered by Adélaïde et al in two pancreatic cancer cell lines (PaTu I, SUIT-2), indicating that NRG1 breakpoints may be a recurring phenomenon in solid tumors.[27] Subsequent studies on breast cancer, pancreatic cancer, and lung cancer tumor samples further emphasized the role of NRG1 rearrangements in tumor development.[28] Comprehensive molecular detection techniques have revealed NRG1 fusions in various other tumors, particularly in invasive mucinous lung adenocarcinoma (IMA) and KRAS wild type pancreatic ductal adenocarcinoma.[29] [30] [31] [32] The identification of recurrent and potentially targetable NRG1 fusions provides therapeutic opportunities for these tumors.

In addition to NRG1 gene fusion, CD74-NRG2 gene fusion has been detected in lung adenocarcinoma patients. NRG2 has moderate affinity with ERBB2/4 heterodimers, and phosphorylation of ERBB2/3/4 may serve as an alternative biomarker for pathway activation.[33] Immunohistochemical analysis of CD74-NRG2 samples showed moderate phosphorylation of ERBB4 in positive tumor cells, whereas EGFR, ERBB2, and ERBB3 did not show phosphorylation. On the other hand, ERBB family members were phosphorylated in NRG1 fusion tumor cells, suggesting that ERBB4 inhibitors may be effective drugs for NRG2 gene fusion tumors.[4]


#
#

Epidemiology of NRG1/2 Gene Fusion in Solid Tumors

Mutation Frequency of NRG1/2 Fusion

The occurrence rate of NRG1 and NRG2 gene fusion in solid tumors is extremely rare. The overall mutation frequency of NRG1 gene fusion in all solid tumors is approximately 0.2%, but in certain patient subgroups, the mutation frequency can be as high as 30%. A study in the United States found an occurrence rate of NRG1 gene fusion of 0.19% among 21,858 cases of solid tumors. The most common tumor types with NRG1 gene fusion are gallbladder cancer, pancreatic cancer, renal cell carcinoma, ovarian cancer, nonsmall cell lung cancer (NSCLC), breast cancer, sarcoma, and bladder cancer. The incidence rates of other tumor types are all less than 0.1%.[3] Data from a population of solid tumor patients in Korea showed an occurrence rate of NRG1 gene fusion of 0.27%, with lung cancer being the most common tumor type.[34] Another study based on data from 13,089 cases of NSCLC in China showed an occurrence rate of NRG1 gene fusion of 0.19%.[35] IMA accounts for approximately 57 to 61% of NRG1 fusion NSCLC and slightly more than half of NRG1 fusion NSCLC patients have never smoked.[36] [37]

The breakpoints of NRG1 fusion are typically found in three specific intronic regions: (1) a 47-kb region between exon 1 and exon 2; (2) a 955-kb region between exon II and exon 2; (3) a region between exon 5 and exon 6, including exon III, with a length of 111 kb.[36] The occurrence rate of NRG2 fusion is even rarer, with a frequency 5 to 10 times lower than that of NRG1.[4] [5] [38]


#

Fusion Partners of NRG1 Gene Fusion

NRG1 gene fusion can have different partners, which affects the biological properties of the synthesized chimeric protein. The NRG1 protein has a domain similar to EGF and acts as a ligand for ERBB3. The ligand can be localized in the complex, while the partner provides a transmembrane domain that binds the ligand to the membrane. In most cases, the partner facilitates the interaction between the ligand and the ERBB3 protein on adjacent cells. CD74 and SLC3A2 are the most common upstream fusion partners, but other partner genes include ATP1B1, CDH1, CLU, CRADD, FUT10, INCENP, KIF22, RBPMS, SLC20A2, VWA8, and XKR6, among others.[34]


#

Other Molecular Characteristics of NRG1 Gene Fusion

Multiple studies have consistently shown that NRG1 gene fusions are generally mutually exclusive with driver genes such as EGFR, ALK, and ROS1. This indicates that NRG1 gene fusion may act as a strong driver mutation promoting the occurrence and development of tumors. Co-occurring mutations with NRG1 gene fusions include TP53 (54.5%), KRAS, BRAF, PIK3CA, NF1, and NF2, among others.[3] [34] Among 15 patients with solid tumors harboring NRG1 gene fusions, the median tumor mutation burden was 3.9/Mb (range: 1.0–51.20/Mb), and the median microsatellite instability was 1.98% (range: 1.0–5.0%).[34]

We believe that NRG1 and NRG2 gene fusions are rare but important targetable oncogenic alterations. Ideally, all advanced and metastatic solid tumors should be systematically tested for NRG1 and NRG2 gene fusions, along with other actionable oncogenic drivers. Molecular testing should be performed at the time of diagnosis, especially for patients with a histopathological diagnosis of IMA. Considering the frequent breakpoints in the intronic region of the NRG1 gene, it is crucial to include intronic coverage when selecting the testing method, especially gene sequencing.


#
#

Detection of NRG1/2 Fusion

Chromosomal translocation is the primary cause of fusion genes, and accurate diagnosis of fusion genes is essential for effective treatment. In the clinical translation of NRG fusion α and β subtypes, it is crucial to avoid false negatives and minimize the need for further confirmation testing due to the diversity and rarity of NRG fusion variants. This requires advanced testing technology with high sensitivity. The standardization of operating procedures can improve the accuracy of detection.[38] Additionally, considering the limited availability of resources in many countries, cost-effectiveness is also an important factor to consider in the testing method. To optimize screening, specific tumor samples and knowledge of NRG fusions in specific cancer types should be combined. Combining multiple testing methods can further enhance the accuracy and reliability of NRG1 fusion detection.

Immunohistochemistry

Immunohistochemistry (IHC) can indirectly detect the fusion status of NRG1 and NRG2 by detecting the protein expression levels of NRG1 or NRG2 and their fusion partners in tumor tissues. IHC has advantages such as fast turnaround time, low cost, high sensitivity, and strong specificity. It relies on specific antibodies that can identify fusion proteins in tumor tissues. However, the selection of antibodies can significantly impact the results, and not all fusion variants may be detectable by specific antibodies.

Indirect detection of pERBB3 immunostaining may serve as a powerful predictive marker for NRG1 fusion, as NRG1 fusion can lead to increased fusion products and chimeric ligands, resulting in ERBB2/ERBB3 heterodimerization and phosphorylation-mediated activation of the ERBB3 receptor.[26] In a study cohort of 85 Caucasian patients, NRG1 rearrangements were investigated in 51 IMA patients and 34 non-IMA patients using NRG1 fluorescence in situ hybridization (FISH), pERBB3 immunohistochemistry, and RNA target sequencing. The findings revealed that 31% of IMA and 3% of non-IMA patients had NRG1 gene rearrangements, indicating that pERBB3 immunohistochemistry had a sensitivity of 94% and specificity of 100% in the 51 IMA samples, as well as a sensitivity of 100% and specificity of 94% in the 34 non-IMA adenocarcinoma samples. Additionally, CD74-NRG1 fusion transcripts were detected in 4 NRG1-positive IMA patients. Importantly, all IMA cases with abnormal pERBB3 expression exhibited NRG1 gene rearrangement.[39] Furthermore, in a study involving 245 lung adenocarcinoma samples, pERBB3 immunohistochemical detection demonstrated a sensitivity of 100% and specificity of 97.5%.[26] Thus, pERBB3 immunohistochemical detection may serve as a rapid and effective prescreening method for identifying NRG1-positive patients.


#

Fluorescence in Situ Hybridization

FISH is a widely utilized method for visualizing and confirming the presence of NRG1 and NRG2 fusions in paraffin-embedded tissue samples. This technique employs fluorescently labeled probes that specifically bind to the fusion genes, enabling precise localization and assessment of fusion events. FISH is particularly valuable in identifying the specific fusion partners and breakpoints involved. When there is a suspicion of NRG1 or NRG2 fusion with distinct characteristics, FISH can be employed for genotyping purposes. Break-apart FISH, a commonly employed clinical method and one of the Food and Drug Administration (FDA)-approved techniques for detecting ALK rearrangements, detects gene fusions. However, unlike ALK fusion FISH testing, the scoring criteria for determining NRG1 fusion positivity lack comprehensive study and validation. Consequently, the current criteria for NRG1 FISH testing positivity temporarily adopt the 15% separation signal threshold used in ALK testing, pending favorable validation data for widespread adoption of NRG1 FISH.[38] While FISH testing has demonstrated success in NSCLC,[40] it was unable to detect NRG1 fusions in two out of three cases of KRAS wild type pancreatic ductal adenocarcinoma with complex NRG1 rearrangement patterns.[31] In addition to its inability to detect complex rearrangement patterns, FISH has other limitations, such as the restricted ability to simultaneously test multiple targets and the inability to determine if fusion partners express fusion products or if other co-mutations are present. Therefore, due to its high cost, low sensitivity, and specificity, we do not recommend FISH as a routine screening method for NRG1 fusion detection.


#

RNA-Based Next-Generation Sequencing

Transcriptome sequencing using second-generation sequencing technology enables accurate identification of NRG1 and NRG2 fusions by comparing gene expression profiles between tumor and normal tissues. This method provides comprehensive information about fusion transcripts and can detect new fusion events. RNA-based next-generation sequencing (NGS) is the optimal tool for discovering fusion genes at the transcriptional level due to the chimeric nature of fusion transcripts. The frequency of NRG1 or NRG2 fusions can be calculated using the number of connected reads, including the β/α isoform ratio. However, RNA-based NGS has limitations in obtaining sufficient quality and quantity of RNA from clinical samples, especially formalin-fixed paraffin-embedded tissues. In the eNRGy1 clinical trial, a combination of DNA and/or RNA NGS and FISH was employed to identify NRG1 fusions. The detection rate of NRG1 fusion using RNA-based NGS was found to be 74% (81/110), whereas the detection rate using DNA-based NGS was only 26%. This highlights the superior advantages of RNA-based NGS in fusion detection.[37]

Whole Transcriptome Sequencing

Whole transcriptome sequencing (WTS) is the most comprehensive method for detecting gene fusions, particularly in identifying new fusion partners. WTS directly sequences transcribed mRNA without relying on initial adapter ligation steps.[30] [31] [40] [41] Unlike targeted RNA sequencing, WTS does not require prior knowledge of fusion partners. However, WTS has limitations such as high requirements for sample quality and quantity, complex data analysis, high cost, and difficulty in detecting low-frequency events.


#

Targeted RNA-Sequencing Panel

Targeted RNA sequencing technology, such as anchored multiplex polymerase chain reaction (AMP), evaluates specific gene expression, mutations, and fusions and improves sequencing coverage by analyzing multiple genes in a single assay.[42] [43] AMP is commercially available but mainly targets genes like ALK, RET, and ROS1 and covers the NRG1 gene.[44] However, it cannot reliably detect NRG2 gene fusions due to the lack of specific primers for NRG2 gene amplification, which is a disadvantage compared with WTS.[3] [38]


#
#

DNA-Based Next-Generation Sequencing

DNA-based NGS technology is widely used for tumor and plasma gene typing. It is a high-throughput sequencing method that provides comprehensive genetic information with reduced costs and time. Hybrid capture technology, a commonly used method, enables the sequencing of translocation breakpoints. DNA-NGS technology can identify most NRG1 gene fusions and determine their breakpoints. However, it may miss fusions with large introns and cannot determine fusion protein functionality. Therefore, we recommend using a DNA gene testing panel that covers the intronic regions of NRG1 and NRG2 genes.


#

Reverse Transcription-Polymerase Chain Reaction

Reverse transcription-polymerase chain reaction is a reliable method for detecting fusion transcripts of NRG1 and NRG2 genes. It involves reverse transcription of RNA into cDNA, followed by amplification using fusion gene-specific primers. This method accurately detects fusion breakpoints and is commonly used for validation, especially for partner genes with a high fusion breakpoint occurrence rate. However, it is not suitable for identifying new fusion partners and may not be sensitive enough for low-abundance fusion transcripts.[45] Therefore, it is not included in our recommended screening strategy.


#

Screening Recommendations for NRG1/2 Fusion

Despite advancements in detection methods, challenges remain in identifying NRG1 and NRG2 gene fusions. These include difficulties in detecting low-abundance fusion transcripts, the need for high-quality samples, lack of standardized methods, and low sensitivity for rare fusion events in heterogeneous tumors.

To enhance the identification of NRG1 gene fusion solid tumor patients, we recommend using DNA or RNA NGS panels targeting the intronic regions of NRG1/2, or pERBB3 immunohistochemistry as the primary screening strategy. RNA NGS technology is particularly recommended when histology and molecular subtypes are unclear. Specific detection strategies and workflow information were listed as follow ([Table 1]; [Fig. 2]).

Zoom Image
Fig. 2 The recommended procedure for the diagnosis and treatment of NRG1/2 gene fusion solid tumors. CTC, circulating tumor cells; IHC, immunohistochemistry; IMA, invasive mucinous adenocarcinoma; NGS, next-generation sequencing; WES, whole exome sequencing; WTS, whole transcriptome sequencing.
Table 1

Consensus on the diagnosis and treatment of NRG1/2 gene fusion solid tumors

Consensus no.

Key points

Recommendation level

Detection time point

Consensus 1

A NRG1/2 gene fusion test, in parallel to other actionable oncogenic drivers' tests is recommended for every adult and pediatric patient with advanced or metastatic solid tumor at diagnosis. NGS testing contain NRG1/2 gene fusions is strongly recommended for invasive mucinous lung adenocarcinoma confirmed by histopathology

Strongly recommended

Consensus 2

Advance or metastatic adult and pediatric solid tumor patients should consider NRG1/2 gene fusion testing before or during standard treatment (recommended). For locally advanced invasive mucinous lung adenocarcinoma patients with high incidence of NRG1/2 gene fusion, a NRG1/2 gene fusion testing is strongly recommended before neoadjuvant therapy

Strongly recommended

Detection method

Consensus 3

Preferred tumor histological specimens should be used for fusion gene testing. If sufficient tumor histological specimens cannot be obtained, cytological specimens may be selected. Prior to fusion gene testing, tissue or cytological specimens should be evaluated for tumor cell content by professional pathologists. If sufficient tumor histological or cytological specimens cannot be obtained, liquid biopsy is recommended as a supplementary testing method

Strongly recommended

Consensus 4

The main methods for NRG1/2 gene fusion testing are whole transcriptome sequencing (WTS), RNA-based NGS panels, and DNA-based NGS panels covering the intronic regions of NRG1/2. The selection of testing platforms and methods should be made reasonably based on sample type, tumor cell content, specimen quality, platform accessibility, testing turnaround time, and cost. RNA-based NGS panels have higher sensitivity than DNA-based NGS panels. If necessary, multiple platforms can be used for complementation and verification, especially when IHC results are positive and DNA-based NGS panel results are negative. In such cases, it is strongly recommended to use the third detection method, RNA-based NGS panel, for confirmation

Strongly recommended

Consensus 5

pERBB3 immunohistochemical test may serve as a rapid and effective prescreening method for identifying NRG1 fusion patients

Strongly recommended

Detection strategy

Consensus 6

Each hospital should establish standardized procedures for NRG1/2 gene fusion testing. Our expert group will regularly issue recommendations on the importance of NGS testing for each type of cancer. Given the rapid development of precision medicine for tumors, promoting the development of precision medicine at different levels of hospitals with NRG1/2 gene fusion testing as a benchmark is of great significance

Recommended

Detection quality control

Consensus 7

All testing should be conducted in accredited laboratories. It is recommended to select laboratories accredited by authoritative institutions such as ISO15189, CAP, and CLIA for testing. The laboratory should perform internal and external quality control related to NRG1/2 gene fusion testing in accordance with relevant regulations

Strongly recommended

Consensus 8

In addition to basic information and quality control data, the testing report should also include tumor cell content, microscopic anatomical status, DNA extraction concentration, and purity. For NGS testing reports, positive fusion results should include chromosome breakpoint positions, participation of tyrosine kinase structural domains, and in-frame fusion data. NRG1/2 gene fusion involving the tyrosine kinase structural domain and being in-frame fusion should be reported as fusion; otherwise, it should be reported as rearrangement

Strongly recommended

Consensus 9

When a physician has doubt, such as inconsistent results from different tests, new partner gene or fusion patterns, complex fusion events, unconfirmed involvement of fusion within the framework or full tyrosine kinase domain, and multiple driver gene positives, etc. discussing results and future treatment decisions with the Molecular Tumor Board (MTB) is strongly recommended

Strongly recommended

Treatment strategy

Consensus 10

For NRG1/2 gene fusion solid tumors, if standard treatment fails, it is recommended to participation in related clinical trials for pan-ERBB TKIs such as afatinib and tarloxotinib, or ERBB2 inhibitory mAbs, ERBB3 inhibitory mAbs or dual anti-ERBB2/ERBB3 mAbs such as zenocutuzumab. For NRG1/2 gene fusion tumor patients with drug resistance, performing NGS test to identify resistance mechanisms and deciding whether other related clinical trials are appropriate is recommended

Recommended

Abbreviations: mAb, monoclonal antibody; NGS, next-generation sequencing; TKI, tyrosine kinase inhibitor.



#
#

Treatment Strategies for NRG1/NRG2 Fusion

Currently, there are no approved targeted therapies specifically for the treatment of NRG1 and NRG2 fusions. However, several potential treatment strategies are being investigated in clinical trials. These include targeting NRG1 fusion solid tumors using TKIs, monoclonal antibodies, or immunotherapy. Due to the intricate molecular pathways associated with NRG1 fusion malignancies, novel therapeutic approaches that target specific mutations or signaling pathways have shown promise in preclinical studies and are currently being evaluated in clinical trials ([Table 2]).

Table 2

Drugs under development for target NRG1 fusion locally advanced or metastatic solid tumors (clinicaltrials.gov accessed on August 1, 2023)

Drug

Target

Developer

Study title

ClinicalTrials.gov ID

Cancer type

Phase

Status

Afatinib

Pan-ERBB TKIs

Boehringer Ingelheim

Afatinib in advanced NRG1-rearranged malignancies: the NCT/DKTK PMO-1604 phase-II trial

NCT04410653

Metastatic and locally advanced NRG1-rearranged malignancies

II

Active, not recruiting

An open-labeled, single-arm clinical study to evaluate the efficacy of afatinib in treatment of locally advanced or metastatic non-small cell lung cancer with NRG1 fusion

NCT04814056

NRG1-fused nonsmall cell lung cancer

IV

Not yet recruiting

Seribantumab

Anti-ERBB3 monoclonal antibody

Merrimack Pharmaceuticals

CRESTONE: a phase 2 study of seribantumab in adult patients with Neuregulin-1 (NRG1) fusion positive locally advanced or metastatic solid tumors

NCT04383210

Locally advanced or metastatic solid tumors

II

Active, not recruiting

Single patient protocol for an NRG1 fusion positive metastatic pancreatic cancer patient using seribantumab

NCT04790695

Metastatic pancreatic cancer

II

Completed

HMBD-001

Anti-ERBB3 monoclonal antibody

Hummingbird Bioscience

A phase 1b study to evaluate HMBD-001 with or without chemotherapy in participants with advanced solid tumors harboring NRG1 gene fusions

NCT05919537

Nonsmall cell lung cancer

Pancreatic cancer

Locally advanced solid tumor

Metastatic solid tumor

I/II

Not yet recruiting

Zenocutuzumab

Anti-ERBB2/ERBB3 monoclonal antibodies

Merus

A phase I/II study of MCLA-128, a full length IgG1 bispecific antibody targeting HER2 and HER3, in patients with solid tumors (eNRGy)

NCT02912949

Solid tumors harboring NRG1 fusion

II

Recruiting

Treatment plan of the HER2/HER3 bispecific antibody, MCLA-128, for a patient with advanced NRG1 fusion positive solid tumor

NCT04100694

Solid tumor

I

Available

A phase 2 study evaluating activity of zenocutuzumab (MCLA-128) in patients with or without molecularly defined cancers

NCT05588609

NSCLC harboring NRG1 fusion

Metastatic castration-resistant prostate cancer

II

Recruiting

Abbreviations: NSCLC, nonsmall cell lung cancer; TKI, tyrosine kinase inhibitor.


Pan-ERBB Tyrosine Kinase Inhibitors

There are several clinical targeted approaches for the treatment of NRG1 and NRG2 fusion tumors, with the inhibition of the ERBB2–ERBB3 heterodimer activity being considered the most effective method.


#

ERBB2 Selective Inhibitor

Afatinib

Afatinib, a pan-ERBB small molecule TKI, irreversibly inhibits tyrosine kinase autophosphorylation by binding to the kinase domains of EGFR, ERBB2, and ERBB4, leading to downregulation of the ERBB signaling. A case series report[46] included six cases of metastatic NRG1 fusion tumors treated with afatinib, comprising five cases of metastatic lung cancer (two mucinous adenocarcinoma and three nonmucinous adenocarcinoma) and one case of metastatic colorectal cancer. Among these cases, one patient with IMA carrying CD74-NRG1 fusion achieved partial remission for over 18 months after treatment with afatinib. Two patients with nonmucinous adenocarcinoma showed sustained responses for over 24 months. One patient with invasive lung mucinous adenocarcinoma carrying SDC4-NRG1 fusion initially achieved partial remission for 5 months with afatinib (40 mg/d), but later experienced lung progression. After increasing the dose of afatinib to 50 mg/d, the patient achieved another 6 months of partial remission. Additionally, one patient with metastatic colorectal cancer carrying POMK-NRG1 fusion and positive KRAS mutation achieved disease stability for 16 months with second-line treatment of afatinib.[46] An alliance composed of 22 centers from 9 countries in Europe, Asia, and the United States provided data on pathologically confirmed NRG1 fusion lung cancer patients, showing an overall response rate (ORR) of 25% for afatinib, independent of the NRG1 fusion subtype, and a median progression-free survival of 2.8 months.[37] Based on these study results, afatinib may be a treatment option for NRG1 fusion tumors.


#

Tarloxotinib

Tarloxotinib is a prodrug that undergoes cleavage under hypoxic conditions to release an effective and irreversible pan-ERBB inhibitor. It represents a novel therapeutic approach that targets the tumor-specific hypoxic environment for cancer treatment. In the MDA-MB-175vIII breast cancer cell line harboring DOC4-NRG1 fusion, tarloxotinib-E effectively inhibits the phosphorylation of ERBB2 and ERBB3 at concentrations similar to afatinib, while simultaneously suppressing the pERK1/2 and pAKT signals.[47] The Phase II RAIN-701 trial, which investigates the use of tarloxotinib as a monotherapy, includes a treatment arm targeting NRG1 fusion tumors (NCT03805841). At present, the results of this subset have not been disclosed.[48]


#
#

ERBB3 Selective Inhibitor

Seribantumab (MM-121, FTN-001)

Seribantumab is a fully human anti-ERBB3 IgG2 monoclonal antibody. Preclinical experiments have shown that seribantumab inhibits the activation of ERBB3 signaling in cells carrying NRG1 gene fusions and disrupts the stability of the entire ERBB family signaling pathway, including the activation of ERBB2, EGFR, and ERBB4.[49] Results from an ongoing Phase II clinical trial, CRESTONE (NCT04383210), evaluating the use of seribantumab in NRG1 fusion-positive solid tumors, demonstrated an ORR of 33% across all cancer types, including two complete responses and a disease control rate of 92%.[50]


#

Lumretuzumab

Lumretuzumab, a polyethylene glycol-engineered humanized monoclonal antibody developed by Roche, aims to inhibit the activation and signal transduction of ERBB3.[51] In cellular experiments using SLC3A2-NRG1 fusion-positive HEK293T cells, lumretuzumab can inhibit the formation of ERBB2/ERBB3 heterocomplex induced by SLC3A2-NRG1 fusion, thereby suppressing the activation of the PI3K/ERK/mTOR signaling pathway and the proliferation and growth of tumor cells.[52]


#
#

ERBB2/ERBB3 Selective Bispecific Monoclonal Antibodies

The ERBB2/ERBB3 bispecific monoclonal antibody, known as zenocutuzumab, targets both ERBB2 and ERBB3 receptors. By doing so, it effectively blocks the activation of ERBB3 by NRG1 fusion protein and inhibits the formation of heterodimers between ERBB2 and ERBB3. This mechanism of action has shown significant efficacy in patients with NRG1 fusion.


#

Zenocutuzumab (MCLA-128)

Zenocutuzumab is a bispecific human IgG1 antibody that contains two separate Fab arms specifically targeting the extracellular domains of ERBB2 and ERBB3. It can simultaneously inhibit the interaction between ERBB2 and NRG1, as well as the heterodimerization between ERBB3 and EGFR. This dual inhibition prevents ERBB3 and ERBB2 heterodimerization.[53] In a clinical trial involving NRG1 fusion-positive/estrogen receptor-positive breast cancer patients who had experienced disease progression after treatment with cyclin-dependent kinase 4/6 inhibitors, zenocutuzumab demonstrated sustained tumor regression.[54] The I/II phase eNRGy clinical trial (NCT02912949) included patients with NRG1 fusions in three cohorts: NSCLC (25 cases), pancreatic cancer (13 cases), and other solid tumors (13 cases). The results of the study showed excellent efficacy of zenocutuzumab in pancreatic cancer patients, with a partial response observed in 42% (5/12) of patients, stable disease in 6 cases, and disease progression in only 1 case. The objective response rate assessed by the researchers in pancreatic cancer was 40% (4/10). In three cases of chemotherapy-resistant NRG1 fusion-positive pancreatic cancer patients, two patients experienced significant tumor shrinkage and sustained benefit for over 12 months: one patient with ATP1B1-NRG1 gene fusion had a 44% reduction in tumor diameter at week 8 of treatment and a 54% reduction after 5 months of treatment, whereas another patient had a 22% reduction in tumor diameter at week 6 of treatment. In a case of CD74-NRG1-positive NSCLC patient who had previously received six systemic treatments including afatinib but experienced rapid disease progression, partial response was achieved for 7 months after switching to zenocutuzumab.[55] Targeting both ERBB2 and ERBB3 simultaneously with zenocutuzumab represents a new treatment approach for NRG1 fusion-positive cancer patients. Based on this, in July 2020, the FDA granted orphan drug designation to zenocutuzumab for the treatment of NRG1 fusion-positive pancreatic cancer patients.


#

Drug Resistance

NRG1 fusion has been identified as a potential mechanism of resistance to targeted therapies. For example, in breast cancer cell lines treated with lapatinib, increased expression of NRG1 has been associated with acquired resistance to EGFR and ERBB2 kinase inhibitors. Overexpression of NRG1 leads to reactivation of EGFR, ERBB2, and ERBB3 through phosphorylation. However, the combination of pertuzumab and lapatinib can inhibit NRG1-induced signaling more effectively than either drug alone. In animal models, this combination therapy has shown greater tumor regression compared with single-drug treatments.[56] Similarly, in selective inhibitors of nuclear export (SINE)-resistant ovarian cancer cell lines, the NRG1/ERBB3 pathway is upregulated. The antitumor effect of SINE can be restored by removing ERBB3 using siRNA.[57] Additionally, exogenous NRG1 can reduce the antitumor effect of SINE in ovarian cancer cell lines with high ERBB3 expression. In ALK-rearranged lung cancer, activation of the NRG1-ERBB3 axis can cause resistance to lorlatinib.[58] However, pharmacological inhibition of ERBB3 or knockdown of the ERBB3 gene can restore sensitivity to lorlatinib in lung cancer cell lines. These findings suggest that targeting the NRG1/ERBB3 axis may be a potential treatment option for resistant cancers. However, it is important to consider the ecological balance between ERBB receptors, as NRG1 can bind to different receptors and unrestricted activation of other ligand–receptor axes may contribute to resistance. Therefore, future drug selection should aim to comprehensively inhibit the ERBB family signaling.[38]


#
#

Summary and Prospect

Tumor-driven fusion protein targets are highly valuable in targeted drug research. The significance of NRG1 fusion in carcinogenesis was initially recognized in the mid-2010s, despite being first reported in breast cancer cell lines in 1997. The recent discovery of NRG2 fusion further emphasizes its importance.

To detect fusion variants of NRG1 and NRG2 genes, particularly in their intronic regions, we propose RNA-based NGS technology, specifically WTS, as the optimal method. Comprehensive molecular profiling analysis of NRG1 and NRG2 fusion solid tumor patients can then identify potential therapeutic targets and guide personalized treatment strategies. This analysis can be achieved through NGS and other advanced genomic technologies. Alternatively, in cases where this is not feasible, IHC detection of pERBB3 levels can serve as a cost-effective preliminary screening method for NRG1 fusion.

Understanding the molecular mechanisms and signaling pathways affecting NRG1 and NRG2 fusion genes is crucial for developing effective treatment strategies. Targeted therapies against these gene variants and signaling pathways have shown promising results in preclinical studies and early clinical trials. Drugs targeting the binding of NRG1 to ERBB3 and/or the heterodimerization of ERBB2/ERBB3, such as the bispecific monoclonal antibody zenocutuzumab, have demonstrated tumor volume reduction in NRG1 fusion-positive tumors. These findings confirm that NRG1 and NRG2 gene fusions, although rare in solid tumors, are actionable oncogenic mutations. Patients who are NRG1 positive and have failed standard treatment are recommended to participate in relevant clinical trials to increase their chances of benefiting.

In conclusion, the management of NRG1 and NRG2 fusion solid tumors necessitates a multidisciplinary approach that encompasses molecular detection methods, targeted therapies, and the selection of combination therapies. Further research and clinical trials are warranted to explore the most effective strategies for addressing these intricate malignancies.


#
#

Conflict of Interest

None declared.

Authors' Contributions

J.C. and Z.S. participated in the design of the expert consensus. C.X., Q.W., D.W., W.W., and W.F. conceived of the expert consensus and participated in its design and other authors coordination and helped to draft the expert consensus. All authors read and approved the final manuscript.


* These authors contributed equally.


  • References

  • 1 Blume-Jensen P, Hunter T. Oncogenic kinase signalling. Nature 2001; 411 (6835) 355-365
  • 2 Falls DL. Neuregulins: functions, forms, and signaling strategies. Exp Cell Res 2003; 284 (01) 14-30
  • 3 Jonna S, Feldman RA, Swensen J. et al. Detection of NRG1 gene fusions in solid tumors. Clin Cancer Res 2019; 25 (16) 4966-4972
  • 4 Kohsaka S, Hayashi T, Nagano M. et al. Identification of novel CD74-NRG2α fusion from comprehensive profiling of lung adenocarcinoma in Japanese never or light smokers. J Thorac Oncol 2020; 15 (06) 948-961
  • 5 Ou SI, Xiu J, Nagasaka M. et al. Identification of novel CDH1-NRG2α and F11R-NRG2α fusions in NSCLC plus additional novel NRG2α fusions in other solid tumors by whole transcriptome sequencing. JTO Clin Res Rep 2020; 2 (02) 100132
  • 6 Odiete O, Hill MF, Sawyer DB. Neuregulin in cardiovascular development and disease. Circ Res 2012; 111 (10) 1376-1385
  • 7 Wilson KJ, Mill CP, Cameron EM, Hobbs SS, Hammer RP, Riese II DJ. Inter-conversion of neuregulin2 full and partial agonists for ErbB4. Biochem Biophys Res Commun 2007; 364 (02) 351-357
  • 8 Hayes NV, Gullick WJ. The neuregulin family of genes and their multiple splice variants in breast cancer. J Mammary Gland Biol Neoplasia 2008; 13 (02) 205-214
  • 9 Holmes WE, Sliwkowski MX, Akita RW. et al. Identification of heregulin, a specific activator of p185erbB2. Science 1992; 256 (5060) 1205-1210
  • 10 Wen D, Peles E, Cupples R. et al. Neu differentiation factor: a transmembrane glycoprotein containing an EGF domain and an immunoglobulin homology unit. Cell 1992; 69 (03) 559-572
  • 11 Marchionni MA, Goodearl AD, Chen MS. et al. Glial growth factors are alternatively spliced erbB2 ligands expressed in the nervous system. Nature 1993; 362 (6418) 312-318
  • 12 Falls DL, Rosen KM, Corfas G, Lane WS, Fischbach GD. ARIA, a protein that stimulates acetylcholine receptor synthesis, is a member of the neu ligand family. Cell 1993; 72 (05) 801-815
  • 13 Tzahar E, Levkowitz G, Karunagaran D. et al. ErbB-3 and ErbB-4 function as the respective low and high affinity receptors of all Neu differentiation factor/heregulin isoforms. J Biol Chem 1994; 269 (40) 25226-25233
  • 14 Hanker AB, Brown BP, Meiler J. et al. Co-occurring gain-of-function mutations in HER2 and HER3 modulate HER2/HER3 activation, oncogenesis, and HER2 inhibitor sensitivity. Cancer Cell 2021; 39 (08) 1099-1114.e8
  • 15 Olayioye MA, Neve RM, Lane HA, Hynes NE. The ErbB signaling network: receptor heterodimerization in development and cancer. EMBO J 2000; 19 (13) 3159-3167
  • 16 Busfield SJ, Michnick DA, Chickering TW. et al. Characterization of a neuregulin-related gene, Don-1, that is highly expressed in restricted regions of the cerebellum and hippocampus. Mol Cell Biol 1997; 17 (07) 4007-4014
  • 17 Higashiyama S, Horikawa M, Yamada K. et al. A novel brain-derived member of the epidermal growth factor family that interacts with ErbB3 and ErbB4. J Biochem 1997; 122 (03) 675-680
  • 18 Carraway III KL, Weber JL, Unger MJ. et al. Neuregulin-2, a new ligand of ErbB3/ErbB4-receptor tyrosine kinases. Nature 1997; 387 (6632) 512-516
  • 19 Hobbs SS, Coffing SL, Le AT. et al. Neuregulin isoforms exhibit distinct patterns of ErbB family receptor activation. Oncogene 2002; 21 (55) 8442-8452
  • 20 Breuleux M. Role of heregulin in human cancer. Cell Mol Life Sci 2007; 64 (18) 2358-2377
  • 21 Forster JA, Paul AB, Harnden P, Knowles MA. Expression of NRG1 and its receptors in human bladder cancer. Br J Cancer 2011; 104 (07) 1135-1143
  • 22 Meyer D, Birchmeier C. Multiple essential functions of neuregulin in development. Nature 1995; 378 (6555) 386-390
  • 23 Schaefer G, Fitzpatrick VD, Sliwkowski MX. Gamma-heregulin: a novel heregulin isoform that is an autocrine growth factor for the human breast cancer cell line, MDA-MB-175. Oncogene 1997; 15 (12) 1385-1394
  • 24 Fernandez-Cuesta L, Thomas RK. Molecular pathways: targeting NRG1 fusions in lung cancer. Clin Cancer Res 2015; 21 (09) 1989-1994
  • 25 Werr L, Plenker D, Dammert MA. et al. CD74-NRG1 fusions are oncogenic in vivo and induce therapeutically tractable ERBB2:ERBB3 heterodimerization. Mol Cancer Ther 2022; 21 (05) 821-830
  • 26 Fernandez-Cuesta L, Plenker D, Osada H. et al. CD74-NRG1 fusions in lung adenocarcinoma. Cancer Discov 2014; 4 (04) 415-422
  • 27 Adélaïde J, Huang HE, Murati A. et al. A recurrent chromosome translocation breakpoint in breast and pancreatic cancer cell lines targets the neuregulin/NRG1 gene. Genes Chromosomes Cancer 2003; 37 (04) 333-345
  • 28 Huang HE, Chin SF, Ginestier C. et al. A recurrent chromosome breakpoint in breast cancer at the NRG1/neuregulin 1/heregulin gene. Cancer Res 2004; 64 (19) 6840-6844
  • 29 Duruisseaux M, McLeer-Florin A, Antoine M. et al. NRG1 fusion in a French cohort of invasive mucinous lung adenocarcinoma. Cancer Med 2016; 5 (12) 3579-3585
  • 30 Jung Y, Yong S, Kim P. et al. VAMP2-NRG1 fusion gene is a novel oncogenic driver of non-small-cell lung adenocarcinoma. J Thorac Oncol 2015; 10 (07) 1107-1111
  • 31 Jones MR, Williamson LM, Topham JT. et al. NRG1 gene fusions are recurrent, clinically actionable gene rearrangements in KRAS wild-type pancreatic ductal adenocarcinoma. Clin Cancer Res 2019; 25 (15) 4674-4681
  • 32 Heining C, Horak P, Uhrig S. et al. NRG1 fusions in KRAS wild-type pancreatic cancer. Cancer Discov 2018; 8 (09) 1087-1095
  • 33 Jones JT, Akita RW, Sliwkowski MX. Binding specificities and affinities of EGF domains for ErbB receptors. FEBS Lett 1999; 447 (2-3): 227-231
  • 34 Cha YJ, Lee C, Joo B, Kim KA, Lee CK, Shim HS. Clinicopathological characteristics of NRG1 fusion-positive solid tumors in Korean patients. Cancer Res Treat 2023; 55 (04) 1087-1095
  • 35 Yuan HCS, Wang L, Dong X, Wang A, Wang K. The landscape of NRG1 fusions based on NGS in Chinese solid tumor patients. ASCO 2022; x: e15073
  • 36 Nagasaka M, Ou SI. Neuregulin 1 fusion-positive NSCLC. J Thorac Oncol 2019; 14 (08) 1354-1359
  • 37 Drilon A, Duruisseaux M, Han JY. et al. Clinicopathologic features and response to therapy of NRG1 fusion-driven lung cancers: the eNRGy1 Global Multicenter Registry. J Clin Oncol 2021; 39 (25) 2791-2802
  • 38 Nagasaka M, Ou SI. NRG1 and NRG2 fusion positive solid tumor malignancies: a paradigm of ligand-fusion oncogenesis. Trends Cancer 2022; 8 (03) 242-258
  • 39 Trombetta D, Graziano P, Scarpa A. et al. Frequent NRG1 fusions in Caucasian pulmonary mucinous adenocarcinoma predicted by phospho-ErbB3 expression. Oncotarget 2018; 9 (11) 9661-9671
  • 40 Jones MR, Lim H, Shen Y. et al. Successful targeting of the NRG1 pathway indicates novel treatment strategy for metastatic cancer. Ann Oncol 2017; 28 (12) 3092-3097
  • 41 Howarth KD, Mirza T, Cooke SL. et al. NRG1 fusions in breast cancer. Breast Cancer Res 2021; 23 (01) 3
  • 42 Mercer TR, Gerhardt DJ, Dinger ME. et al. Targeted RNA sequencing reveals the deep complexity of the human transcriptome. Nat Biotechnol 2011; 30 (01) 99-104
  • 43 Song Z, Xu C, He Y. et al. Simultaneous detection of gene fusions and base mutations in cancer tissue biopsies by sequencing dual nucleic acid templates in unified reaction. Clin Chem 2020; 66 (01) 178-187
  • 44 Zheng Z, Liebers M, Zhelyazkova B. et al. Anchored multiplex PCR for targeted next-generation sequencing. Nat Med 2014; 20 (12) 1479-1484
  • 45 Lanic MD, Le Loarer F, Rainville V. et al. Detection of sarcoma fusions by a next-generation sequencing based-ligation-dependent multiplex RT-PCR assay. Mod Pathol 2022; 35 (05) 649-663
  • 46 Cadranel J, Liu SV, Duruisseaux M. et al. Therapeutic potential of afatinib in NRG1 fusion-driven solid tumors: a case series. Oncologist 2021; 26 (01) 7-16
  • 47 Estrada-Bernal A, Le AT, Doak AE. et al. Tarloxotinib is a hypoxia-activated Pan-HER kinase inhibitor active against a broad range of HER-family oncogenes. Clin Cancer Res 2021; 27 (05) 1463-1475
  • 48 Liu SV. NRG1 fusions: biology to therapy. Lung Cancer 2021; 158: 25-28
  • 49 Odintsov I, Lui AJW, Sisso WJ. et al. The anti-HER3 mAb seribantumab effectively inhibits growth of patient-derived and isogenic cell line and xenograft models with oncogenic NRG1 fusions. Clin Cancer Res 2021; 27 (11) 3154-3166
  • 50 Thavaneswaran S, Chan WY, Asghari R. et al. Clinical response to seribantumab, an anti-human epidermal growth factor receptor-3 immunoglobulin 2 monoclonal antibody, in a patient with metastatic pancreatic ductal adenocarcinoma harboring an NRG1 fusion. JCO Precis Oncol 2022; 6: e2200263
  • 51 Meneses-Lorente G, Friess T, Kolm I. et al. Preclinical pharmacokinetics, pharmacodynamics, and efficacy of RG7116: a novel humanized, glycoengineered anti-HER3 antibody. Cancer Chemother Pharmacol 2015; 75 (04) 837-850
  • 52 Shin DH, Jo JY, Han JY. Dual targeting of ERBB2/ERBB3 for the treatment of SLC3A2-NRG1-mediated lung cancer. Mol Cancer Ther 2018; 17 (09) 2024-2033
  • 53 Geuijen CAW, De Nardis C, Maussang D. et al. Unbiased combinatorial screening identifies a bispecific IgG1 that potently inhibits HER3 signaling via HER2-guided ligand blockade. Cancer Cell 2021; 39 (08) 1163-1164
  • 54 Fontana E, Torga G, Fostea R. et al. Sustained tumor regression with zenocutuzumab, a bispecific antibody targeting human epidermal growth factor receptor 2/human epidermal growth factor receptor 3 signaling, in NRG1 fusion-positive, estrogen receptor-positive breast cancer after progression on a cyclin-dependent kinase 4/6 inhibitor. JCO Precis Oncol 2022; 6: e2100446
  • 55 Schram AM, Odintsov I, Espinosa-Cotton M. et al. Zenocutuzumab, a HER2xHER3 bispecific antibody, is effective therapy for tumors driven by NRG1 gene rearrangements. Cancer Discov 2022; 12 (05) 1233-1247
  • 56 Leung WY, Roxanis I, Sheldon H. et al. Combining lapatinib and pertuzumab to overcome lapatinib resistance due to NRG1-mediated signalling in HER2-amplified breast cancer. Oncotarget 2015; 6 (08) 5678-5694
  • 57 Miyake TM, Pradeep S, Bayraktar E. et al. NRG1/ERBB3 pathway activation induces acquired resistance to XPO1 inhibitors. Mol Cancer Ther 2020; 19 (08) 1727-1735
  • 58 Taniguchi H, Akagi K, Dotsu Y. et al. Pan-HER inhibitors overcome lorlatinib resistance caused by NRG1/HER3 activation in ALK-rearranged lung cancer. Cancer Sci 2023; 114 (01) 164-173

Address for correspondence

Zheng-bo Song, MD, PhD
Department of Chemotherapy, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital)
Hangzhou, Zhejiang 310022
People's Republic of China   
Jing Chen, MD, PhD
Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
Wuhan Hubei 430022
People's Republic of China   

Publication History

Article published online:
27 February 2024

© 2024. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Blume-Jensen P, Hunter T. Oncogenic kinase signalling. Nature 2001; 411 (6835) 355-365
  • 2 Falls DL. Neuregulins: functions, forms, and signaling strategies. Exp Cell Res 2003; 284 (01) 14-30
  • 3 Jonna S, Feldman RA, Swensen J. et al. Detection of NRG1 gene fusions in solid tumors. Clin Cancer Res 2019; 25 (16) 4966-4972
  • 4 Kohsaka S, Hayashi T, Nagano M. et al. Identification of novel CD74-NRG2α fusion from comprehensive profiling of lung adenocarcinoma in Japanese never or light smokers. J Thorac Oncol 2020; 15 (06) 948-961
  • 5 Ou SI, Xiu J, Nagasaka M. et al. Identification of novel CDH1-NRG2α and F11R-NRG2α fusions in NSCLC plus additional novel NRG2α fusions in other solid tumors by whole transcriptome sequencing. JTO Clin Res Rep 2020; 2 (02) 100132
  • 6 Odiete O, Hill MF, Sawyer DB. Neuregulin in cardiovascular development and disease. Circ Res 2012; 111 (10) 1376-1385
  • 7 Wilson KJ, Mill CP, Cameron EM, Hobbs SS, Hammer RP, Riese II DJ. Inter-conversion of neuregulin2 full and partial agonists for ErbB4. Biochem Biophys Res Commun 2007; 364 (02) 351-357
  • 8 Hayes NV, Gullick WJ. The neuregulin family of genes and their multiple splice variants in breast cancer. J Mammary Gland Biol Neoplasia 2008; 13 (02) 205-214
  • 9 Holmes WE, Sliwkowski MX, Akita RW. et al. Identification of heregulin, a specific activator of p185erbB2. Science 1992; 256 (5060) 1205-1210
  • 10 Wen D, Peles E, Cupples R. et al. Neu differentiation factor: a transmembrane glycoprotein containing an EGF domain and an immunoglobulin homology unit. Cell 1992; 69 (03) 559-572
  • 11 Marchionni MA, Goodearl AD, Chen MS. et al. Glial growth factors are alternatively spliced erbB2 ligands expressed in the nervous system. Nature 1993; 362 (6418) 312-318
  • 12 Falls DL, Rosen KM, Corfas G, Lane WS, Fischbach GD. ARIA, a protein that stimulates acetylcholine receptor synthesis, is a member of the neu ligand family. Cell 1993; 72 (05) 801-815
  • 13 Tzahar E, Levkowitz G, Karunagaran D. et al. ErbB-3 and ErbB-4 function as the respective low and high affinity receptors of all Neu differentiation factor/heregulin isoforms. J Biol Chem 1994; 269 (40) 25226-25233
  • 14 Hanker AB, Brown BP, Meiler J. et al. Co-occurring gain-of-function mutations in HER2 and HER3 modulate HER2/HER3 activation, oncogenesis, and HER2 inhibitor sensitivity. Cancer Cell 2021; 39 (08) 1099-1114.e8
  • 15 Olayioye MA, Neve RM, Lane HA, Hynes NE. The ErbB signaling network: receptor heterodimerization in development and cancer. EMBO J 2000; 19 (13) 3159-3167
  • 16 Busfield SJ, Michnick DA, Chickering TW. et al. Characterization of a neuregulin-related gene, Don-1, that is highly expressed in restricted regions of the cerebellum and hippocampus. Mol Cell Biol 1997; 17 (07) 4007-4014
  • 17 Higashiyama S, Horikawa M, Yamada K. et al. A novel brain-derived member of the epidermal growth factor family that interacts with ErbB3 and ErbB4. J Biochem 1997; 122 (03) 675-680
  • 18 Carraway III KL, Weber JL, Unger MJ. et al. Neuregulin-2, a new ligand of ErbB3/ErbB4-receptor tyrosine kinases. Nature 1997; 387 (6632) 512-516
  • 19 Hobbs SS, Coffing SL, Le AT. et al. Neuregulin isoforms exhibit distinct patterns of ErbB family receptor activation. Oncogene 2002; 21 (55) 8442-8452
  • 20 Breuleux M. Role of heregulin in human cancer. Cell Mol Life Sci 2007; 64 (18) 2358-2377
  • 21 Forster JA, Paul AB, Harnden P, Knowles MA. Expression of NRG1 and its receptors in human bladder cancer. Br J Cancer 2011; 104 (07) 1135-1143
  • 22 Meyer D, Birchmeier C. Multiple essential functions of neuregulin in development. Nature 1995; 378 (6555) 386-390
  • 23 Schaefer G, Fitzpatrick VD, Sliwkowski MX. Gamma-heregulin: a novel heregulin isoform that is an autocrine growth factor for the human breast cancer cell line, MDA-MB-175. Oncogene 1997; 15 (12) 1385-1394
  • 24 Fernandez-Cuesta L, Thomas RK. Molecular pathways: targeting NRG1 fusions in lung cancer. Clin Cancer Res 2015; 21 (09) 1989-1994
  • 25 Werr L, Plenker D, Dammert MA. et al. CD74-NRG1 fusions are oncogenic in vivo and induce therapeutically tractable ERBB2:ERBB3 heterodimerization. Mol Cancer Ther 2022; 21 (05) 821-830
  • 26 Fernandez-Cuesta L, Plenker D, Osada H. et al. CD74-NRG1 fusions in lung adenocarcinoma. Cancer Discov 2014; 4 (04) 415-422
  • 27 Adélaïde J, Huang HE, Murati A. et al. A recurrent chromosome translocation breakpoint in breast and pancreatic cancer cell lines targets the neuregulin/NRG1 gene. Genes Chromosomes Cancer 2003; 37 (04) 333-345
  • 28 Huang HE, Chin SF, Ginestier C. et al. A recurrent chromosome breakpoint in breast cancer at the NRG1/neuregulin 1/heregulin gene. Cancer Res 2004; 64 (19) 6840-6844
  • 29 Duruisseaux M, McLeer-Florin A, Antoine M. et al. NRG1 fusion in a French cohort of invasive mucinous lung adenocarcinoma. Cancer Med 2016; 5 (12) 3579-3585
  • 30 Jung Y, Yong S, Kim P. et al. VAMP2-NRG1 fusion gene is a novel oncogenic driver of non-small-cell lung adenocarcinoma. J Thorac Oncol 2015; 10 (07) 1107-1111
  • 31 Jones MR, Williamson LM, Topham JT. et al. NRG1 gene fusions are recurrent, clinically actionable gene rearrangements in KRAS wild-type pancreatic ductal adenocarcinoma. Clin Cancer Res 2019; 25 (15) 4674-4681
  • 32 Heining C, Horak P, Uhrig S. et al. NRG1 fusions in KRAS wild-type pancreatic cancer. Cancer Discov 2018; 8 (09) 1087-1095
  • 33 Jones JT, Akita RW, Sliwkowski MX. Binding specificities and affinities of EGF domains for ErbB receptors. FEBS Lett 1999; 447 (2-3): 227-231
  • 34 Cha YJ, Lee C, Joo B, Kim KA, Lee CK, Shim HS. Clinicopathological characteristics of NRG1 fusion-positive solid tumors in Korean patients. Cancer Res Treat 2023; 55 (04) 1087-1095
  • 35 Yuan HCS, Wang L, Dong X, Wang A, Wang K. The landscape of NRG1 fusions based on NGS in Chinese solid tumor patients. ASCO 2022; x: e15073
  • 36 Nagasaka M, Ou SI. Neuregulin 1 fusion-positive NSCLC. J Thorac Oncol 2019; 14 (08) 1354-1359
  • 37 Drilon A, Duruisseaux M, Han JY. et al. Clinicopathologic features and response to therapy of NRG1 fusion-driven lung cancers: the eNRGy1 Global Multicenter Registry. J Clin Oncol 2021; 39 (25) 2791-2802
  • 38 Nagasaka M, Ou SI. NRG1 and NRG2 fusion positive solid tumor malignancies: a paradigm of ligand-fusion oncogenesis. Trends Cancer 2022; 8 (03) 242-258
  • 39 Trombetta D, Graziano P, Scarpa A. et al. Frequent NRG1 fusions in Caucasian pulmonary mucinous adenocarcinoma predicted by phospho-ErbB3 expression. Oncotarget 2018; 9 (11) 9661-9671
  • 40 Jones MR, Lim H, Shen Y. et al. Successful targeting of the NRG1 pathway indicates novel treatment strategy for metastatic cancer. Ann Oncol 2017; 28 (12) 3092-3097
  • 41 Howarth KD, Mirza T, Cooke SL. et al. NRG1 fusions in breast cancer. Breast Cancer Res 2021; 23 (01) 3
  • 42 Mercer TR, Gerhardt DJ, Dinger ME. et al. Targeted RNA sequencing reveals the deep complexity of the human transcriptome. Nat Biotechnol 2011; 30 (01) 99-104
  • 43 Song Z, Xu C, He Y. et al. Simultaneous detection of gene fusions and base mutations in cancer tissue biopsies by sequencing dual nucleic acid templates in unified reaction. Clin Chem 2020; 66 (01) 178-187
  • 44 Zheng Z, Liebers M, Zhelyazkova B. et al. Anchored multiplex PCR for targeted next-generation sequencing. Nat Med 2014; 20 (12) 1479-1484
  • 45 Lanic MD, Le Loarer F, Rainville V. et al. Detection of sarcoma fusions by a next-generation sequencing based-ligation-dependent multiplex RT-PCR assay. Mod Pathol 2022; 35 (05) 649-663
  • 46 Cadranel J, Liu SV, Duruisseaux M. et al. Therapeutic potential of afatinib in NRG1 fusion-driven solid tumors: a case series. Oncologist 2021; 26 (01) 7-16
  • 47 Estrada-Bernal A, Le AT, Doak AE. et al. Tarloxotinib is a hypoxia-activated Pan-HER kinase inhibitor active against a broad range of HER-family oncogenes. Clin Cancer Res 2021; 27 (05) 1463-1475
  • 48 Liu SV. NRG1 fusions: biology to therapy. Lung Cancer 2021; 158: 25-28
  • 49 Odintsov I, Lui AJW, Sisso WJ. et al. The anti-HER3 mAb seribantumab effectively inhibits growth of patient-derived and isogenic cell line and xenograft models with oncogenic NRG1 fusions. Clin Cancer Res 2021; 27 (11) 3154-3166
  • 50 Thavaneswaran S, Chan WY, Asghari R. et al. Clinical response to seribantumab, an anti-human epidermal growth factor receptor-3 immunoglobulin 2 monoclonal antibody, in a patient with metastatic pancreatic ductal adenocarcinoma harboring an NRG1 fusion. JCO Precis Oncol 2022; 6: e2200263
  • 51 Meneses-Lorente G, Friess T, Kolm I. et al. Preclinical pharmacokinetics, pharmacodynamics, and efficacy of RG7116: a novel humanized, glycoengineered anti-HER3 antibody. Cancer Chemother Pharmacol 2015; 75 (04) 837-850
  • 52 Shin DH, Jo JY, Han JY. Dual targeting of ERBB2/ERBB3 for the treatment of SLC3A2-NRG1-mediated lung cancer. Mol Cancer Ther 2018; 17 (09) 2024-2033
  • 53 Geuijen CAW, De Nardis C, Maussang D. et al. Unbiased combinatorial screening identifies a bispecific IgG1 that potently inhibits HER3 signaling via HER2-guided ligand blockade. Cancer Cell 2021; 39 (08) 1163-1164
  • 54 Fontana E, Torga G, Fostea R. et al. Sustained tumor regression with zenocutuzumab, a bispecific antibody targeting human epidermal growth factor receptor 2/human epidermal growth factor receptor 3 signaling, in NRG1 fusion-positive, estrogen receptor-positive breast cancer after progression on a cyclin-dependent kinase 4/6 inhibitor. JCO Precis Oncol 2022; 6: e2100446
  • 55 Schram AM, Odintsov I, Espinosa-Cotton M. et al. Zenocutuzumab, a HER2xHER3 bispecific antibody, is effective therapy for tumors driven by NRG1 gene rearrangements. Cancer Discov 2022; 12 (05) 1233-1247
  • 56 Leung WY, Roxanis I, Sheldon H. et al. Combining lapatinib and pertuzumab to overcome lapatinib resistance due to NRG1-mediated signalling in HER2-amplified breast cancer. Oncotarget 2015; 6 (08) 5678-5694
  • 57 Miyake TM, Pradeep S, Bayraktar E. et al. NRG1/ERBB3 pathway activation induces acquired resistance to XPO1 inhibitors. Mol Cancer Ther 2020; 19 (08) 1727-1735
  • 58 Taniguchi H, Akagi K, Dotsu Y. et al. Pan-HER inhibitors overcome lorlatinib resistance caused by NRG1/HER3 activation in ALK-rearranged lung cancer. Cancer Sci 2023; 114 (01) 164-173

Zoom Image
Fig. 1 NRG1 and NRG2 structures. (A) NRG1 possesses I, II, and III subtype structures patterns. The coding sequences of the same isoform vary due to diverse transcription start sites and alternative splicing of NRG1 gene promoters. It is worth noting that the EGF-like domain alone has the capability to efficiently activate homologous ERBB receptor tyrosine kinases. N and C marked in red represent the N-terminal and C-terminal of NRG1 protein, respectively. To obtain further information, please refer to the relevant literature.[2] [38] (B) NRG2 structure. CRD, cysteine-rich domain; CTc, cytoplasmic tail domain C terminal of the EGF-like domain; TMc, transmembrane domain C terminal of the EGF-like domain; TMn, transmembrane domain N-terminal of the EGF-like domain.
Zoom Image
Fig. 2 The recommended procedure for the diagnosis and treatment of NRG1/2 gene fusion solid tumors. CTC, circulating tumor cells; IHC, immunohistochemistry; IMA, invasive mucinous adenocarcinoma; NGS, next-generation sequencing; WES, whole exome sequencing; WTS, whole transcriptome sequencing.