Next Article in Journal
Preoperative and Intraoperative Identification of Sentinel Lymph Nodes in Melanoma Surgery
Previous Article in Journal
Discrimination of Lung Cancer and Benign Lung Diseases Using BALF Exosome DNA Methylation Profile
Previous Article in Special Issue
Baseline Blood CD8+ T Cell Activation Potency Discriminates Responders from Non-Responders to Immune Checkpoint Inhibition Combined with Stereotactic Radiotherapy in Non-Small-Cell Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

NRG1 Gene Fusions—What Promise Remains Behind These Rare Genetic Alterations? A Comprehensive Review of Biology, Diagnostic Approaches, and Clinical Implications

1
Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-059 Lublin, Poland
2
Department of Zoology and Nature Conservation, Institute of Biology, Maria Curie-Sklodowska University in Lublin, 20-033 Lublin, Poland
3
Oncology Clinic, Institute of the Polish Mother’s Health Center in Lodz, 93-338 Lodz, Poland
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(15), 2766; https://doi.org/10.3390/cancers16152766
Submission received: 11 July 2024 / Revised: 1 August 2024 / Accepted: 3 August 2024 / Published: 5 August 2024
(This article belongs to the Special Issue Novel Biomarkers in Non-small Cell Lung Cancer (NSCLC))

Abstract

:

Simple Summary

Neuregulin-1 (NRG1) is an important regulator of ErbB-mediated pathways involved in cancer development. Recently, there have been several studies analyzing NRG-1 gene fusions engaged in altering the dimerization of HER family proteins and the consecutive results of their activation in different types of cancer. Non-small cell lung cancer (NSCLC) patients can benefit from pan-HER inhibitors, and knowledge of NRG-1 fusions can help tailor the treatment to a specific group of patients. New drugs targeting cells with NRG-1 fusions are under clinical trials and show effectiveness in NSCLC treatment.

Abstract

Non-small cell lung cancer (NSCLC) presents a variety of druggable genetic alterations that revolutionized the treatment approaches. However, identifying new alterations may broaden the group of patients benefitting from such novel treatment options. Recently, the interest focused on the neuregulin-1 gene (NRG1), whose fusions may have become a potential predictive factor. To date, the occurrence of NRG1 fusions has been considered a negative prognostic marker in NSCLC treatment; however, many premises remain behind the targetability of signaling pathways affected by the NRG1 gene. The role of NRG1 fusions in ErbB-mediated cell proliferation especially seems to be considered as a main target of treatment. Hence, NSCLC patients harboring NRG1 fusions may benefit from targeted therapies such as pan-HER family inhibitors, which have shown efficacy in previous studies in various cancers, and anti-HER monoclonal antibodies. Considering the increased interest in the NRG1 gene as a potential clinical target, in the following review, we highlight its biology, as well as the potential clinical implications that were evaluated in clinics or remained under consideration in clinical trials.

1. Introduction

In the era of precision therapy, novel driver alterations are extensively studied to qualify the patient for the best-fitting treatment. In 2020, lung cancer accounted for 11.9% of all new cancer diagnoses in Europe, constituting about 480,000 people [1]. Non-small cell lung cancer (NSCLC), which includes 85% of lung cancer cases, is one of the cancer types that presents a variety of actionable genetic changes, with quite a few available targeted drugs that have revolutionized the treatment approaches [2]. Nevertheless, the percentage of patients that receive already popular epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), anaplastic lymphoma kinase (ALK) inhibitors, or less common molecules is still low. The search for new targets is still ongoing to allow a larger group of patients treatment tailored specifically for them [3]. Identifying such alterations or fusions may effectively select those benefiting from such novel treatment options. Apart from gene mutations, rearrangements and fusions of different genes are among the most commonly diagnosed cancer cell driver alterations [2]. The inhibitors of anaplastic lymphoma kinase (ALK), ROS proto-oncogene 1 (ROS1), rearranged during transfection (RET), and neurotrophic tyrosine receptor kinase (NTRK) rearrangements are already present in our everyday clinical practice [4,5]. Recently, interest has focused on neuregulin-1 (NRG1) as a potential oncogenic target.
The NRG1 gene harbors several variants that have been classified on the Evidence for Sequence-variant Classification (ESCAT) scale as likely benign (rs3924999—intron region; rs7832768—promoter region) or uncertain significance (rs10503929 and rs16879552—both intronic), and their clinical relevance should be confirmed [6]. On the other hand, NRG1 fusions might be considered the main oncogenic factor in solid tumors. The first description of such fusions (CD74-NRG1) in invasive mucinous adenocarcinoma of the lung (IMA) was in 2014. The targeted drugs for patients with NRG1 fusions are still in clinical trials, and the search continues [7].
NRG1 rearrangements are uncommon compared to other more often described gene alterations found in NSCLC. In one of the studies, it was present in 0.5% of patients (2 of 404 analyzed cases) [8], in the other in 0.3% of patients (25 of 9252 analyzed samples) [9]. The prevalence of NRG1 rearrangements in other types of cancers is similar to NSCLC and amounts to 0.5% in cholangiocarcinoma, pancreatic carcinoma, and renal cell carcinoma, 0.4% in ovarian cancer, and 0.2% in breast cancer and sarcoma [7,9,10]. Thus, the NRG1 fusions may be considered as biomarkers in various cancer types. Moreover, these fusions exclude the occurrence of other cancer-driving genetic changes. In some NSCLC cases, however, their presence was described along with mutations in KRAS and BRAF or ALK rearrangements [9,11].
To date, the occurrence of NRG1 fusions was considered a negative prognostic marker in NSCLC treatment. Patients harboring such alterations presented reduced overall survival (OS) when treated with standard chemotherapy, chemoimmunotherapy, or immunotherapy alone. However, there are many premises behind the targetability of pathways affected by this alteration [9,12,13].
Considering the increased interest in the NRG1 gene as a potential clinical target, this review discusses the structure and biology of the NRG1 gene and the occurrence of potential genetic fusions. Furthermore, we indicate the NRG1 fusion detection methods that are based on both high-throughput or single-gene approaches. In the end, we highlight the druggable applications of NRG1 fusions as the first or secondary target in the treatment of NSCLC, which are already available in clinics or are still under consideration in clinical trials.

2. Structure and Biology of NRG1 Fusions

Neuregulin 1 is a protein, encoded by the NRG1 gene located on the short arm of chromosome 8 (8p12), that is involved in various biological processes, including neural development, synaptic plasticity, myelination, and inter-cell signaling in the heart and breast [14]. The function of NRG1 is necessary for the early stages of development, and its absence, as was shown in mouse models, does not allow for proper embryonic development [15]. NRG1 gene has many tissue-specific isoforms, created through alternative splicing, that differ structurally from each other. However, most isoforms contain the same extracellular epidermal growth factor-like (EGF-like) domain [14,16,17], which is crucial in the case of NRG1 fusions to keep the functionality of the aberrant protein and drive cancer cell development. Most isoforms of NRG1 are bound to the cell membrane as a precursor. During proteolytic processes, the mature NRG1 is released, which can be transported further from the cell of origin and activate receptors on the surface of other cells. However, isoform III of neuregulin 1 retains the EGF-like domain in the membrane, which allows for the activation of mainly neighboring cells [18]. Moreover, there are some premises that epigenetic changes may also dysregulate the NRG1 expression, leading to its involvement in cancer development and progression [19]. The schematic localisation and structure of the NRG1 gene is presented in Figure 1.
The EGF-like domain of NRG1 protein is mainly an activator of Erb-B2 tyrosine kinase receptor 3 (ErbB3 also called HER3, human epidermal growth factor receptor 3), subsequently activating heterodimerization, most frequently ErbB2-ErbB3, but also EGFR or ErbB4, and further downstream signaling through mitogen-activated protein kinase (MAPK) and phosphoinositide-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathways [20,21]. Although ErbB3 has seriously decreased kinase activity, its dimerization with other ErbB family receptors, after activation by NRG1, allows further downstream activation of the aforementioned pathways [22,23]. The NRG1 fusion proteins act as abnormal activators of ErbB-mediated cell proliferation pathways, and the result of such activation is the promotion of proliferation of molecularly altered cells.
It is postulated that NRG1 fusions act similarly to neuregulin-1 isoform III, with a membrane-attached EGF-like domain. Different NRG1 fusions can activate different homo- or heterodimers of ErbB [24], hence, they can activate diverse downstream pathways and result in alternative results from blockade attempts. The scheme of dimeric ErbB downstream signaling pathways regulated by the NRG1 protein is presented in Figure 2.

3. Occurrence of NRG1 Fusions

As previously stated, the first discovery of the CD74-NRG1 fusion was described in 2014 in a study of 25 lung adenocarcinoma patients without KRAS or EGFR mutations. The described five cases were detected in non-smoking females with the IMA subtype [7]. Since that discovery, most of the CD74-NRG1 fusion cases have been presented in this subtype of lung adenocarcinoma [25]. Subsequently, other groups of researchers identified different fusion partners of the NRG1 gene: SLC3A2 [9,12,26], SDC4 [9,27], RBPMS [9,28], VAMP2 [29], WRN [28], ATP1B1 [27], ROCK1 [25], RALGAPA1 [30], TNC [9], MDK [9], DIP2B [9], MRPL13 [9], DPYSL2 [9], PARP8 [9], THAP7 [25], SMAD4 [25], KIF13B [13], ITGB1 [31], UBXN8 [10], NPTN [32], CADM1 [33], F11R [33], FGFR1 [33], FLYWCH1 [33], KRAS [33], PLCG2 [33], and VAPB [33]. To date, the study by Jonna and co-workers is the most comprehensive analysis regarding NRG1 fusions in solid tumors, where the incidence of the most common partners was as follows: CD74 (29%), ATP1B1 (10%), SDC4 (7%), and RBPMS (5%). All the other fusions detected in the analyzed group of 21,858 solid tumor samples occurred with 2% frequency [9].
Fusions of NRG1 and a few different partner genes have been described in other tumors as well, namely in ovarian cancer: SETD4 [9], TSHZ2 [9], ZMYM2 [9], RAB3IL1 [25], and CLU [25,34], and in pancreatic ductal adenocarcinoma: VTCN1 [9], CDH1 [9], CDH6 [35], SARAF [10,35], APP [36], and CDK1 [37]. Other described individual cases include breast cancer: ADAM9 [9], COX10-AS1 [9], AKAP13 [25], FOXA1 [25], DDHD2 [10], FUT10 [10], BRE [10], CD9 [10], ARHGEF39 [38], FAM91A1 [38], and ZNF704 [38], colorectal carcinoma: IKBKB [10], ZCCHC7 [10], TNRFSF10B [10], ERO1L [10], and KCTD9 [10], esophageal carcinoma: BIN3 [10] and CCAR2 [10], gallbladder carcinoma: NOTCH2 [9], head and neck squamous carcinoma: THBS1 [25] and PDE7A [25], bladder cancer: GDF15 [9], renal cell carcinoma: PCM1 [25], prostate carcinoma: STMN2 [25] and UNC5D [39], neuroendocrine tumor of the nasopharynx: HMBOX1 [9,40], spindle cell sarcoma: WHSC1L1 [9] and PPHLN1 [40], as well as uterine carcinosarcoma: PMEPA1 [25]. Interestingly, in fusions of NRG1 and PCM1, STMN2, and PMEPA1, the EGF-like domain was not observed; hence, the functionality and the activating ability of these fusions may not be relevant as oncogenic drivers [25]. The overview of locations of NRG1 fusion gene partners in different tumors within chromosome 8 and other chromosomes is presented in Table 1 and Figure 3 and Figure 4, respectively.

4. Detection of NRG1 Fusions

The rare occurrence of NRG1 fusions requires a robust detection method, especially in a wide range of partner genes. The main obstacle is to capture all probable fusions in a single sample using an economically viable tool.
The comprehensive identification of NRG1 fusions may involve next-generation sequencing (NGS) technologies, both RNA and DNA-based, which allow for high-throughput genomic profiling of tumor samples [8,41]. Since the gene spans over 1Mb, NGS allows for full analysis of all NRG1 alterations, those known and unknown as well [9]. RNA-based sequencing can be used to identify fusions located in-frame, allowing for detecting products of transcription from alternative splicing forms, which are common in the case of the NRG1 gene [8,9]. The most useful method in such an approach would be whole transcriptome sequencing (WTS), as it allows the detection of all possible transcripts. However, the drawback of WTS is that the method needs high-quality RNA isolated from the sample [28,42]. The DNA-based NGS approaches, whole exome sequencing (WES) and targeted sequencing allow, on the other hand, for the description of exact sequences of breakpoints but do not tell if these sequences undergo translation [9]. Such methods also do not cover intronic sequences properly, which is a disadvantage, as the NRG1 gene consists of large, non-coding fragments that might carry the possible breaking points [16]. Another drawback of the DNA-based NGS approach is the poor quality of DNA extracted from formalin-fixed paraffin-embedded (FFPE) tissue samples that, in the computational analysis, may deliver a high number of artifacts that may imitate the false positive results [43,44]. However, due to the mentioned limitations and high costs of NGS-based approaches, other single gene-based methods are still of great interest for NRG1 gene detection.
The most common technique used for the detection of fusion genes and their protein products is immunohistochemistry (IHC). The method is relatively fast, cheap, and provides high sensitivity and specificity, although it needs a qualified pathologist for proper description [45]. It was postulated that phosphorylated ErbB3 (pErbB3) protein analysis could be the first step to identifying tumors carrying NRG1 fusions [21,33]. The association between high pErbB3 expression detected with IHC in IMA and non-IMA lung cancer samples was shown by Trombetta et al. [46]. The main issue with IHC is that it might show false-positive results, as it presents fusion proteins that undergo full expression (transcription and translation) [47], hence, the method can be used mainly as the first step in screening and selection of samples for further, more complex analysis [9].
The third approach to the detection of NRG1 fusions is the fluorescence in situ hybridization (FISH) technique [48]. It is commonly available in most molecular laboratories, but it requires more expertise and experience from the diagnostician when interpreting the results. It is more labor-consuming and works well with previously described fusions. Also, the technique cannot describe specific breakage points in fusion partners [36,46]. Besides IHC and FISH techniques, real-time PCR and Sanger sequencing also allow the detection of the exact known genomic breakpoints but remain underused and are very limiting [9]. On the other hand, Nanostring technology may become the RNA-based approach that will allow efficient estimation of the level of expression of all the exons in the region of interest within the NRG1 gene [49].

5. NRG1 Fusion as the Predictive Factor in Lung Cancer Treatment

The activation of signal transduction by binding of NRG1 ligand to ErbB family receptors or the process of ErbB family protein dimerization is considered the main target of treatment in patients harboring NRG1 fusions [50,51]. Hence, NSCLC patients harboring NRG1 fusions may benefit from targeted therapies such as HER family inhibitors, which have shown efficacy in previous studies in various cancers. The first choice in such an approach would be afatinib. This irreversible pan-HER inhibitor was proven effective in NSCLC patients harboring EGFR gene-activating mutations [52,53]. Several studies analyzed the effectiveness of afatinib in patients harboring NRG1 fusions, although they were mainly case studies. Drilon et al. reported no response to afatinib treatment in four patients with IMA histology, although there were visible results in patient-derived xenograft mouse models [25]. Gay et al. presented two cases of lung cancer patients without EGFR mutations, carriers of SLC3A2-NRG1 and CD74-NRG1 fusions. The patients received afatinib with documented durable responses of 10 and 12 months, respectively [8]. Another case study of five lung cancer patients harboring CD74-NRG1 or SDC4-NRG1 fusions, treated with afatinib, resulted in four cases of partial response (PR) (5–27 months) and one stable disease (SD) (4 months) [54]. On the other hand, a single-patient case study with a CD74-NRG1 fusion presented by Wu et al. indicated that afatinib showed PR for seven months until the progression of the disease [55]. A larger study by Liu et al. with different types of tumors included 29 NSCLC patients treated with afatinib, and it showed a 48.3% overall response rate (ORR), including three complete responses (CRs) and eleven PRs, with a median duration of response (DoR) of 6.8 months and median PFS of 6.1 months [56].
Tarloxotinib, another small molecule pan-ErbB inhibitor, in a hypoxic tumor environment decreased the phosphorylated ErbB-related process by targeting the membrane of reductase STEP4 protein, leading to tumor growth inhibition and cancer regression. The results were observed in patient-derived cell lines and multiple murine xenograft models harboring an NRG1 fusion [9,57,58]. Apart from the small-molecule pan-ErbB inhibitors mentioned above, there are also many positive premises behind the inhibition of NRG1-related pathways by monoclonal antibodies binding to the ErbB receptors. Odintsov et al. reported that seribantumab (anti-ErbB3 antibody, MM-121/SAR256212) decreased activation of the PI3K-AKT, mTOR, and ERK pathways in NRG1 fusion-positive patient-derived lung and breast cancer cell lines and patient-derived xenograft (PDX) models from lung and ovarian cancer patients. Moreover, seribantumab efficiently blocked other ErbB family members, indicating a similar to afatinib reduction of proliferation and induction of apoptosis [59]. In the end, Drilon et al. observed durable tumor regression in a PDX mouse model and anti-proliferative activity in the MDA-MB-175-VII cell line [25]. The summary of the effect of different drugs on the prognosis of lung cancer patients is presented in Table 2.

6. NRG1 Fusion as a Secondary Target in Lung Cancer Treatment

NRG1 gene fusions have the potential to affect the activity of ErbB-related pathways; thus, from one side, there is a potential treatment option for cancer patients harboring NRG1 fusion-positive cancers by HER-targeted therapies [60]. However, some studies have indicated that NRG1 fusion drives the primary resistance to molecularly targeted therapies by activation of the HER3 [61] and HER3/AKT [62] signaling pathway. Due to the complexity of the ErbB-related signal transduction pathways, the NRG1-driven resistance has the potential to be overcome by the application of treatment regimens based on multi-targeted agents. For instance, trastuzumab combined with anti-HER3 monoclonal antibody, pertuzumab, or poziotinib may revert the resistance process in cell lines [61,62,63].
In NSCLC, NRG1 fusions are listed as acquired oncogenic alterations associated with the acquired resistance to EGFR-TKIs driven by activation of the NRG1/ErbB3 pathway [64,65]. Moreover, it was also shown that ALK-rearranged NSCLC cells acquire resistance to ALK inhibitors, losing the EML4/ALK fusion and activating the NRG1/ErbB3 pathway [9]. In such a situation, the sensitivity to crizotinib may be restored by pan-ErbB inhibitors, afatinib or dacomitinib, in the absence of other secondary ALK mutations [66]. In case of resistance to alectinib, the NRG1/ErbB3 activation maintains survival and stimulates mesenchymal activity, driving the epithelial–mesenchymal transition (EMT) that is the main hallmark of cancer dissemination [67,68]. This phenomenon may be confirmed by the observation that high expression of ErbB3 and NRG1 significantly correlated with brain metastases from primary lung tumors [69].

7. Clinical Trials Related to Patients with Solid Tumors Harboring NRG1 Fusions

Besides the published results, there are also some clinical trials evaluating the targeted treatment possibilities in patients harboring NRG1 fusions (Table 3). The clinical trial NCT03805841 [70] evaluated the ORR to tarloxotinib in NSCLC patients harboring insertion in exon 20 of the EGFR gene, activating mutation of HER2 or NRG1 fusion. However, the study was terminated, and the outcome has not been provided yet. The efficient blocking of ErbB family members by seribantumab was confirmed in metastatic cancer patients having high and low levels of NRG1 and ErbB2 expression, respectively (NCT01447706 [71], NCT01151046 [72], NCT00994123 [73]). Moreover, in the CRESTONE study (NCT04383210) in the cohort of NSCLC patients harboring NRG1 fusions who received seribantumab, the ORR and the disease control rate were 39% and 94%, respectively. The overall duration of response ranged from 1.4 to 17.2 months [74,75].
Further, Zenocutuzumab (MCLA-128), a bispecific monoclonal antibody against ErbB2 and ErbB3, in the eNRGy study (NCT02912949) demonstrated durable efficacy and a well-tolerated safety profile in patients with advanced solid tumors harboring NRG1 fusion, regardless of tumor histology [76]. Moreover, the NCT01966445 trial showed that GSK2849330, an anti-ErbB3 monoclonal antibody, elicited a durable 19-month response in NSCLC patients harboring the CD74-NRG1 fusion. In the end, poziotinib in the ZENITH20 clinical trial (NCT03318939) demonstrated antitumor activity with a durable response and manageable safety profile as the second-generation TKI in previously treated NSCLC patients with HER2 exon 20 insertions [77,78,79].

8. Conclusions and Future Perspectives

The application of deep sequencing techniques, such as next-generation sequencing, has provided a wide array of data about the molecular background of NSCLC and opened routes for treatment personalization. This advancement revolutionized the management of therapy in these deadly conditions. Moreover, the studies shed light on how rare alterations affect the signaling pathways, indicating they impact treatment response or acquire resistance to targeted approaches. To date, the occurrence of NRG1 fusions, which is a very rare alteration in solid tumors, was considered a negative prognostic marker in NSCLC treatment; however, gaining knowledge about its impact on ErbB signaling pathways has provided significant attention in recent scientific research, offering a potential avenue for targeted therapy. Recent and ongoing clinical trials and preclinical studies have explored the effectiveness of both already available and new agents in NRG1 fusion-positive lung cancers, demonstrating promising results in terms of response rates and disease control. Thus, including such NSCLC cases in planning treatment regimens is reasonable. Moreover, re-evaluating standard approaches to NSCLC molecular analysis to detect possibly actionable, novel gene fusions or alterations that may affect well-known signaling pathways seems relevant and shows promise for further clinical improvement.

Author Contributions

Writing—review and editing, T.K., M.N., M.K. and E.K. Graphics—T.K. and M.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Dyba, T.; Randi, G.; Bray, F.; Martos, C.; Giusti, F.; Nicholson, N.; Gavin, A.; Flego, M.; Neamtiu, L.; Dimitrova, N.; et al. The European Cancer Burden in 2020: Incidence and Mortality Estimates for 40 Countries and 25 Major Cancers. Eur. J. Cancer 2021, 157, 308–347. [Google Scholar] [CrossRef]
  2. Wang, M.; Herbst, R.S.; Boshoff, C. Toward Personalized Treatment Approaches for Non-Small-Cell Lung Cancer. Nat. Med. 2021, 27, 1345–1356. [Google Scholar] [CrossRef]
  3. Xiao, Y.; Liu, P.; Wei, J.; Zhang, X.; Guo, J.; Lin, Y. Recent Progress in Targeted Therapy for Non-Small Cell Lung Cancer. Front. Pharmacol. 2023, 14, 1125547. [Google Scholar] [CrossRef]
  4. Schram, A.M.; Chang, M.T.; Jonsson, P.; Drilon, A. Fusions in Solid Tumours: Diagnostic Strategies, Targeted Therapy, and Acquired Resistance. Nat. Rev. Clin. Oncol. 2017, 14, 735–748. [Google Scholar] [CrossRef]
  5. Chen, J.; Xu, C.; Lv, J.; Lu, W.; Zhang, Y.; Wang, D.; Song, Y. Clinical Characteristics and Targeted Therapy of Different Gene Fusions in Non-Small Cell Lung Cancer: A Narrative Review. Transl. Lung Cancer Res. 2023, 12, 895–908. [Google Scholar] [CrossRef]
  6. Horak, P.; Griffith, M.; Danos, A.M.; Pitel, B.A.; Madhavan, S.; Liu, X.; Chow, C.; Williams, H.; Carmody, L.; Barrow-Laing, L.; et al. Standards for the Classification of Pathogenicity of Somatic Variants in Cancer (Oncogenicity): Joint Recommendations of Clinical Genome Resource (ClinGen), Cancer Genomics Consortium (CGC), and Variant Interpretation for Cancer Consortium (VICC). Genet. Med. Off. J. Am. Coll. Med. Genet. 2022, 24, 986–998. [Google Scholar] [CrossRef]
  7. Fernandez-Cuesta, L.; Plenker, D.; Osada, H.; Sun, R.; Menon, R.; Leenders, F.; Ortiz-Cuaran, S.; Peifer, M.; Bos, M.; Daßler, J.; et al. CD74-NRG1 Fusions in Lung Adenocarcinoma. Cancer Discov. 2014, 4, 415–422. [Google Scholar] [CrossRef]
  8. Gay, N.D.; Wang, Y.; Beadling, C.; Warrick, A.; Neff, T.; Corless, C.L.; Tolba, K. Durable Response to Afatinib in Lung Adenocarcinoma Harboring NRG1 Gene Fusions. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2017, 12, e107–e110. [Google Scholar] [CrossRef]
  9. Jonna, S.; Feldman, R.A.; Swensen, J.; Gatalica, Z.; Korn, W.M.; Borghaei, H.; Ma, P.C.; Nieva, J.J.; Spira, A.I.; Vanderwalde, A.M.; et al. Detection of NRG1 Gene Fusions in Solid Tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 4966–4972. [Google Scholar] [CrossRef]
  10. Severson, E.; Achyut, B.R.; Nesline, M.; Pabla, S.; Previs, R.A.; Kannan, G.; Chenn, A.; Zhang, S.; Klein, R.; Conroy, J.; et al. RNA Sequencing Identifies Novel NRG1 Fusions in Solid Tumors That Lack Co-Occurring Oncogenic Drivers. J. Mol. Diagn. 2023, 25, 454–466. [Google Scholar] [CrossRef]
  11. Muscarella, L.A.; Trombetta, D.; Fabrizio, F.P.; Scarpa, A.; Fazio, V.M.; Maiello, E.; Rossi, A.; Graziano, P. ALK and NRG1 Fusions Coexist in a Patient with Signet Ring Cell Lung Adenocarcinoma. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2017, 12, e161–e163. [Google Scholar] [CrossRef]
  12. Shin, D.H.; Kim, S.H.; Choi, M.; Bae, Y.-K.; Han, C.; Choi, B.K.; Kim, S.S.; Han, J.-Y. Oncogenic KRAS Promotes Growth of Lung Cancer Cells Expressing SLC3A2-NRG1 Fusion via ADAM17-Mediated Shedding of NRG1. Oncogene 2022, 41, 280–292. [Google Scholar] [CrossRef]
  13. Xia, D.; Le, L.P.; Iafrate, A.J.; Lennerz, J. KIF13B-NRG1 Gene Fusion and KRAS Amplification in a Case of Natural Progression of Lung Cancer. Int. J. Surg. Pathol. 2017, 25, 238–240. [Google Scholar] [CrossRef]
  14. Falls, D.L. Neuregulins: Functions, Forms, and Signaling Strategies. Exp. Cell Res. 2003, 284, 14–30. [Google Scholar] [CrossRef]
  15. Liu, S.V. NRG1 Fusions: Biology to Therapy. Lung Cancer Amst. Neth. 2021, 158, 25–28. [Google Scholar] [CrossRef]
  16. Meyer, D.; Yamaai, T.; Garratt, A.; Riethmacher-Sonnenberg, E.; Kane, D.; Theill, L.E.; Birchmeier, C. Isoform-Specific Expression and Function of Neuregulin. Dev. Camb. Engl. 1997, 124, 3575–3586. [Google Scholar] [CrossRef]
  17. Steinthorsdottir, V.; Stefansson, H.; Ghosh, S.; Birgisdottir, B.; Bjornsdottir, S.; Fasquel, A.C.; Olafsson, O.; Stefansson, K.; Gulcher, J.R. Multiple Novel Transcription Initiation Sites for NRG1. Gene 2004, 342, 97–105. [Google Scholar] [CrossRef]
  18. Wang, J.Y.; Miller, S.J.; Falls, D.L. The N-Terminal Region of Neuregulin Isoforms Determines the Accumulation of Cell Surface and Released Neuregulin Ectodomain. J. Biol. Chem. 2001, 276, 2841–2851. [Google Scholar] [CrossRef]
  19. Li, H.; Xu, L.; Cao, H.; Wang, T.; Yang, S.; Tong, Y.; Wang, L.; Liu, Q. Analysis on the Pathogenesis and Treatment Progress of NRG1 Fusion-Positive Non-Small Cell Lung Cancer. Front. Oncol. 2024, 14, 1405380. [Google Scholar] [CrossRef]
  20. Gollamudi, M.; Nethery, D.; Liu, J.; Kern, J.A. Autocrine Activation of ErbB2/ErbB3 Receptor Complex by NRG-1 in Non-Small Cell Lung Cancer Cell Lines. Lung Cancer 2004, 43, 135–143. [Google Scholar] [CrossRef]
  21. Fernandez-Cuesta, L.; Thomas, R.K. Molecular Pathways: Targeting NRG1 Fusions in Lung Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2015, 21, 1989–1994. [Google Scholar] [CrossRef]
  22. Guy, P.M.; Platko, J.V.; Cantley, L.C.; Cerione, R.A.; Carraway, K.L. Insect Cell-Expressed p180erbB3 Possesses an Impaired Tyrosine Kinase Activity. Proc. Natl. Acad. Sci. USA 1994, 91, 8132–8136. [Google Scholar] [CrossRef]
  23. Kim, H.-G.; Lee, C.-K.; Cho, S.-M.; Whang, K.; Cha, B.-H.; Shin, J.-H.; Song, K.-H.; Jeong, S.-W. Neuregulin 1 Up-Regulates the Expression of Nicotinic Acetylcholine Receptors through the ErbB2/ErbB3-PI3K-MAPK Signaling Cascade in Adult Autonomic Ganglion Neurons. J. Neurochem. 2013, 124, 502–513. [Google Scholar] [CrossRef]
  24. Muthuswamy, S.K.; Gilman, M.; Brugge, J.S. Controlled Dimerization of ErbB Receptors Provides Evidence for Differential Signaling by Homo- and Heterodimers. Mol. Cell. Biol. 1999, 19, 6845–6857. [Google Scholar] [CrossRef]
  25. Drilon, A.; Somwar, R.; Mangatt, B.P.; Edgren, H.; Desmeules, P.; Ruusulehto, A.; Smith, R.S.; Delasos, L.; Vojnic, M.; Plodkowski, A.J.; et al. Response to ERBB3-Directed Targeted Therapy in NRG1-Rearranged Cancers. Cancer Discov. 2018, 8, 686–695. [Google Scholar] [CrossRef]
  26. Nakaoku, T.; Tsuta, K.; Ichikawa, H.; Shiraishi, K.; Sakamoto, H.; Enari, M.; Furuta, K.; Shimada, Y.; Ogiwara, H.; Watanabe, S.; et al. Druggable Oncogene Fusions in Invasive Mucinous Lung Adenocarcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014, 20, 3087–3093. [Google Scholar] [CrossRef]
  27. Jones, M.R.; Lim, H.; Shen, Y.; Pleasance, E.; Ch’ng, C.; Reisle, C.; Leelakumari, S.; Zhao, C.; Yip, S.; Ho, J.; et al. Successful Targeting of the NRG1 Pathway Indicates Novel Treatment Strategy for Metastatic Cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2017, 28, 3092–3097. [Google Scholar] [CrossRef]
  28. Dhanasekaran, S.M.; Balbin, O.A.; Chen, G.; Nadal, E.; Kalyana-Sundaram, S.; Pan, J.; Veeneman, B.; Cao, X.; Malik, R.; Vats, P.; et al. Transcriptome Meta-Analysis of Lung Cancer Reveals Recurrent Aberrations in NRG1 and Hippo Pathway Genes. Nat. Commun. 2014, 5, 5893. [Google Scholar] [CrossRef]
  29. Jung, Y.; Yong, S.; Kim, P.; Lee, H.-Y.; Jung, Y.; Keum, J.; Lee, S.; Kim, J.; Kim, J. VAMP2-NRG1 Fusion Gene Is a Novel Oncogenic Driver of Non-Small-Cell Lung Adenocarcinoma. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2015, 10, 1107–1111. [Google Scholar] [CrossRef]
  30. McCoach, C.E.; Le, A.T.; Gowan, K.; Jones, K.; Schubert, L.; Doak, A.; Estrada-Bernal, A.; Davies, K.D.; Merrick, D.T.; Paul, A.; et al. Resistance Mechanisms to Targeted Therapies in ROS1+ and ALK+ Non-Small Cell Lung Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 3334. [Google Scholar] [CrossRef]
  31. Pan, Y.; Zhang, Y.; Ye, T.; Zhao, Y.; Gao, Z.; Yuan, H.; Zheng, D.; Zheng, S.; Li, H.; Li, Y.; et al. Detection of Novel NRG1, EGFR, and MET Fusions in Lung Adenocarcinomas in the Chinese Population. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2019, 14, 2003–2008. [Google Scholar] [CrossRef]
  32. Nie, X.; Zhang, P.; Bie, Z.; Song, C.; Zhang, M.; Ma, D.; Cui, D.; Cheng, G.; Li, H.; Lei, Y.; et al. Durable Response to Afatinib in Advanced Lung Adenocarcinoma Harboring a Novel NPTN-NRG1 Fusion: A Case Report. World J. Surg. Oncol. 2023, 21, 246. [Google Scholar] [CrossRef]
  33. Drilon, A.; Duruisseaux, M.; Han, J.-Y.; Ito, M.; Falcon, C.; Yang, S.-R.; Murciano-Goroff, Y.R.; Chen, H.; Okada, M.; Molina, M.A.; et al. Clinicopathologic Features and Response to Therapy of NRG1 Fusion-Driven Lung Cancers: The eNRGy1 Global Multicenter Registry. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2021, 39, 2791–2802. [Google Scholar] [CrossRef]
  34. Murumägi, A.; Ungureanu, D.; Khan, S.; Hirasawa, A.; Arjama, M.; Välimäki, K.; Mikkonen, P.; Niininen, W.; Kumar, A.; Eldfors, S.; et al. Abstract 2945: Clinical Implementation of Precision Systems Oncology in the Treatment of Ovarian Cancer Based on Ex-Vivo Drug Testing and Molecular Profiling. Cancer Res. 2019, 79, 2945. [Google Scholar] [CrossRef]
  35. Heining, C.; Horak, P.; Uhrig, S.; Codo, P.L.; Klink, B.; Hutter, B.; Fröhlich, M.; Bonekamp, D.; Richter, D.; Steiger, K.; et al. NRG1 Fusions in KRAS Wild-Type Pancreatic Cancer. Cancer Discov. 2018, 8, 1087–1095. [Google Scholar] [CrossRef]
  36. Laskin, J.; Liu, S.V.; Tolba, K.; Heining, C.; Schlenk, R.F.; Cheema, P.; Cadranel, J.; Jones, M.R.; Drilon, A.; Cseh, A.; et al. NRG1 Fusion-Driven Tumors: Biology, Detection, and the Therapeutic Role of Afatinib and Other ErbB-Targeting Agents. Ann. Oncol. 2020, 31, 1693–1703. [Google Scholar] [CrossRef]
  37. Jones, M.R.; Williamson, L.M.; Topham, J.T.; Lee, M.K.C.; Goytain, A.; Ho, J.; Denroche, R.E.; Jang, G.; Pleasance, E.; Shen, Y.; et al. NRG1 Gene Fusions Are Recurrent, Clinically Actionable Gene Rearrangements in KRAS Wild-Type Pancreatic Ductal Adenocarcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 4674–4681. [Google Scholar] [CrossRef]
  38. Howarth, K.D.; Mirza, T.; Cooke, S.L.; Chin, S.-F.; Pole, J.C.; Turro, E.; Eldridge, M.D.; Garcia, R.M.; Rueda, O.M.; Boursnell, C.; et al. NRG1 Fusions in Breast Cancer. Breast Cancer Res. 2021, 23, 3. [Google Scholar] [CrossRef]
  39. Ptáková, N.; Martínek, P.; Holubec, L.; Janovský, V.; Vančurová, J.; Grossmann, P.; Navarro, P.A.; Rodriguez Moreno, J.F.; Alaghehbandan, R.; Hes, O.; et al. Identification of Tumors with NRG1 Rearrangement, Including a Novel Putative Pathogenic UNC5D-NRG1 Gene Fusion in Prostate Cancer by Data-Drilling a de-Identified Tumor Database. Genes. Chromosomes Cancer 2021, 60, 474–481. [Google Scholar] [CrossRef]
  40. Dermawan, J.K.; Zou, Y.; Antonescu, C.R. Neuregulin 1 (NRG1) Fusion-Positive High-Grade Spindle Cell Sarcoma: A Distinct Group of Soft Tissue Tumors with Metastatic Potential. Genes Chromosomes Cancer 2022, 61, 123–130. [Google Scholar] [CrossRef]
  41. Sturgill, E.G.; Srivastava, J.; Correia, J.; Schumacher, C.; Luckett, D.; Perez, C.A.; Wang, J.S.; Divers, S.G.; Bashir, B.; Johnson, J.; et al. Abstract 921: Identification of NRG1 Fusions in Patients with Solid Tumors: Analysis from a Real-World Community Oncology Network. Cancer Res. 2023, 83, 921. [Google Scholar] [CrossRef]
  42. Chang, J.C.; Offin, M.; Falcon, C.; Brown, D.; Houck-Loomis, B.R.; Meng, F.; Rudneva, V.A.; Won, H.H.; Amir, S.; Montecalvo, J.; et al. Comprehensive Molecular and Clinicopathologic Analysis of 200 Pulmonary Invasive Mucinous Adenocarcinomas Identifies Distinct Characteristics of Molecular Subtypes. Clin. Cancer Res. 2021, 27, 4066–4076. [Google Scholar] [CrossRef] [PubMed]
  43. Cappello, F.; Angerilli, V.; Munari, G.; Ceccon, C.; Sabbadin, M.; Pagni, F.; Fusco, N.; Malapelle, U.; Fassan, M. FFPE-Based NGS Approaches into Clinical Practice: The Limits of Glory from a Pathologist Viewpoint. J. Pers. Med. 2022, 12, 750. [Google Scholar] [CrossRef] [PubMed]
  44. Mathieson, W.; Thomas, G.A. Why Formalin-Fixed, Paraffin-Embedded Biospecimens Must Be Used in Genomic Medicine: An Evidence-Based Review and Conclusion. J. Histochem. Cytochem. Off. J. Histochem. Soc. 2020, 68, 543–552. [Google Scholar] [CrossRef]
  45. Dixon, A.R.; Bathany, C.; Tsuei, M.; White, J.; Barald, K.F.; Takayama, S. Recent Developments in Multiplexing Techniques for Immunohistochemistry. Expert Rev. Mol. Diagn. 2015, 15, 1171–1186. [Google Scholar] [CrossRef] [PubMed]
  46. Trombetta, D.; Graziano, P.; Scarpa, A.; Sparaneo, A.; Rossi, G.; Rossi, A.; Di Maio, M.; Antonello, D.; Mafficini, A.; Fabrizio, F.P.; et al. Frequent NRG1 Fusions in Caucasian Pulmonary Mucinous Adenocarcinoma Predicted by Phospho-ErbB3 Expression. Oncotarget 2018, 9, 9661–9671. [Google Scholar] [CrossRef] [PubMed]
  47. O’Hurley, G.; Sjöstedt, E.; Rahman, A.; Li, B.; Kampf, C.; Pontén, F.; Gallagher, W.M.; Lindskog, C. Garbage in, Garbage out: A Critical Evaluation of Strategies Used for Validation of Immunohistochemical Biomarkers. Mol. Oncol. 2014, 8, 783–798. [Google Scholar] [CrossRef] [PubMed]
  48. Adélaïde, J.; Huang, H.-E.; Murati, A.; Alsop, A.E.; Orsetti, B.; Mozziconacci, M.-J.; Popovici, C.; Ginestier, C.; Letessier, A.; Basset, C.; et al. A Recurrent Chromosome Translocation Breakpoint in Breast and Pancreatic Cancer Cell Lines Targets the Neuregulin/NRG1 Gene. Genes Chromosomes Cancer 2003, 37, 333–345. [Google Scholar] [CrossRef] [PubMed]
  49. Alì, G.; Bruno, R.; Savino, M.; Giannini, R.; Pelliccioni, S.; Menghi, M.; Boldrini, L.; Proietti, A.; Chella, A.; Ribechini, A.; et al. Analysis of Fusion Genes by NanoString System: A Role in Lung Cytology? Arch. Pathol. Lab. Med. 2018, 142, 480–489. [Google Scholar] [CrossRef]
  50. Shin, D.H.; Jo, J.Y.; Han, J.-Y. Dual Targeting of ERBB2/ERBB3 for the Treatment of SLC3A2-NRG1-Mediated Lung Cancer. Mol. Cancer Ther. 2018, 17, 2024–2033. [Google Scholar] [CrossRef]
  51. Rosas, D.; Raez, L.E.; Russo, A.; Rolfo, C. Neuregulin 1 Gene (NRG1). A Potentially New Targetable Alteration for the Treatment of Lung Cancer. Cancers 2021, 13, 5038. [Google Scholar] [CrossRef]
  52. Harvey, R.D.; Adams, V.R.; Beardslee, T.; Medina, P. Afatinib for the Treatment of EGFR Mutation-Positive NSCLC: A Review of Clinical Findings. J. Oncol. Pharm. Pract. Off. Publ. Int. Soc. Oncol. Pharm. Pract. 2020, 26, 1461–1474. [Google Scholar] [CrossRef]
  53. Jiang, Y.; Fang, X.; Xiang, Y.; Fang, T.; Liu, J.; Lu, K. Afatinib for the Treatment of NSCLC with Uncommon EGFR Mutations: A Narrative Review. Curr. Oncol. 2023, 30, 5337–5349. [Google Scholar] [CrossRef]
  54. Cadranel, J.; Liu, S.V.; Duruisseaux, M.; Branden, E.; Goto, Y.; Weinberg, B.A.; Heining, C.; Schlenk, R.F.; Cheema, P.; Jones, M.R.; et al. Therapeutic Potential of Afatinib in NRG1 Fusion-Driven Solid Tumors: A Case Series. Oncologist 2021, 26, 7–16. [Google Scholar] [CrossRef]
  55. Wu, X.; Zhang, D.; Shi, M.; Wang, F.; Li, Y.; Lin, Q. Successful Targeting of the NRG1 Fusion Reveals Durable Response to Afatinib in Lung Adenocarcinoma: A Case Report. Ann. Transl. Med. 2021, 9, 1507. [Google Scholar] [CrossRef]
  56. Liu, S.V.; Frohn, C.; Minasi, L.; Fernamberg, K.; Klink, A.J.; Gajra, A.; Savill, K.M.Z.; Jonna, S. Real-World Outcomes Associated with Afatinib Use in Patients with Solid Tumors Harboring NRG1 Gene Fusions. Lung Cancer 2024, 188, 107469. [Google Scholar] [CrossRef]
  57. Bhandari, V.; Hoey, C.; Liu, L.Y.; Lalonde, E.; Ray, J.; Livingstone, J.; Lesurf, R.; Shiah, Y.-J.; Vujcic, T.; Huang, X.; et al. Molecular Landmarks of Tumor Hypoxia across Cancer Types. Nat. Genet. 2019, 51, 308–318. [Google Scholar] [CrossRef]
  58. Estrada-Bernal, A.; Le, A.T.; Doak, A.E.; Tirunagaru, V.G.; Silva, S.; Bull, M.R.; Smaill, J.B.; Patterson, A.V.; Kim, C.; Liu, S.V.; et al. Tarloxotinib Is a Hypoxia-Activated Pan-HER Kinase Inhibitor Active against a Broad Range of HER-Family Oncogenes. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 1463–1475. [Google Scholar] [CrossRef]
  59. Odintsov, I.; Lui, A.J.W.; Sisso, W.J.; Gladstone, E.; Liu, Z.; Delasos, L.; Kurth, R.I.; Sisso, E.M.; Vojnic, M.; Khodos, I.; et al. The Anti-HER3 Monoclonal Antibody Seribantumab Effectively Inhibits Growth of Patient-Derived and Isogenic Cell Line and Xenograft Models with Oncogenic NRG1 Fusions. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 3154–3166. [Google Scholar] [CrossRef]
  60. Swain, S.M.; Shastry, M.; Hamilton, E. Targeting HER2-Positive Breast Cancer: Advances and Future Directions. Nat. Rev. Drug Discov. 2023, 22, 101–126. [Google Scholar] [CrossRef]
  61. Yang, L.; Li, Y.; Shen, E.; Cao, F.; Li, L.; Li, X.; Wang, X.; Kariminia, S.; Chang, B.; Li, H.; et al. NRG1-Dependent Activation of HER3 Induces Primary Resistance to Trastuzumab in HER2-Overexpressing Breast Cancer Cells. Int. J. Oncol. 2017, 51, 1553–1562. [Google Scholar] [CrossRef]
  62. Guardia, C.; Bianchini, G.; Arpí-LLucià, O.; Menendez, S.; Casadevall, D.; Galbardi, B.; Dugo, M.; Servitja, S.; Montero, J.C.; Soria-Jiménez, L.; et al. Preclinical and Clinical Characterization of Fibroblast-Derived Neuregulin-1 on Trastuzumab and Pertuzumab Activity in HER2-Positive Breast Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 5096–5108. [Google Scholar] [CrossRef]
  63. Udagawa, H.; Robichaux, J.P.; Elamin, Y.Y.; He, J.; Nilsson, M.B.; Heymach, J.V. Abstract 1182: Molecular Landscape and ErbB Family Signaling in NRG Fusion NSCLC: Therapeutic Implications for Pan-ErbB Family Inhibitors. Cancer Res. 2022, 82, 1182. [Google Scholar] [CrossRef]
  64. Trombetta, D.; Sparaneo, A.; Fabrizio, F.P.; Di Micco, C.M.; Rossi, A.; Muscarella, L.A. NRG1 and NRG2 Fusions in Non-Small Cell Lung Cancer (NSCLC): Seven Years between Lights and Shadows. Expert Opin. Ther. Targets 2021, 25, 865–875. [Google Scholar] [CrossRef]
  65. Taniguchi, H.; Yamada, T.; Wang, R.; Tanimura, K.; Adachi, Y.; Nishiyama, A.; Tanimoto, A.; Takeuchi, S.; Araujo, L.H.; Boroni, M.; et al. AXL Confers Intrinsic Resistance to Osimertinib and Advances the Emergence of Tolerant Cells. Nat. Commun. 2019, 10, 259. [Google Scholar] [CrossRef]
  66. Taniguchi, H.; Akagi, K.; Dotsu, Y.; Yamada, T.; Ono, S.; Imamura, E.; Gyotoku, H.; Takemoto, S.; Yamaguchi, H.; Sen, T.; et al. Pan-HER Inhibitors Overcome Lorlatinib Resistance Caused by NRG1/HER3 Activation in ALK-Rearranged Lung Cancer. Cancer Sci. 2023, 114, 164–173. [Google Scholar] [CrossRef]
  67. Tanimura, K.; Yamada, T.; Okada, K.; Nakai, K.; Horinaka, M.; Katayama, Y.; Morimoto, K.; Ogura, Y.; Takeda, T.; Shiotsu, S.; et al. HER3 Activation Contributes toward the Emergence of ALK Inhibitor-Tolerant Cells in ALK-Rearranged Lung Cancer with Mesenchymal Features. NPJ Precis. Oncol. 2022, 6, 5. [Google Scholar] [CrossRef]
  68. Isozaki, H.; Ichihara, E.; Takigawa, N.; Ohashi, K.; Ochi, N.; Yasugi, M.; Ninomiya, T.; Yamane, H.; Hotta, K.; Sakai, K.; et al. Non-Small Cell Lung Cancer Cells Acquire Resistance to the ALK Inhibitor Alectinib by Activating Alternative Receptor Tyrosine Kinases. Cancer Res. 2016, 76, 1506–1516. [Google Scholar] [CrossRef]
  69. Saunus, J.M.; Quinn, M.C.J.; Patch, A.-M.; Pearson, J.V.; Bailey, P.J.; Nones, K.; McCart Reed, A.E.; Miller, D.; Wilson, P.J.; Al-Ejeh, F.; et al. Integrated Genomic and Transcriptomic Analysis of Human Brain Metastases Identifies Alterations of Potential Clinical Significance. J. Pathol. 2015, 237, 363–378. [Google Scholar] [CrossRef]
  70. Ye, L.; Chen, X.; Zhou, F. EGFR-Mutant NSCLC: Emerging Novel Drugs. Curr. Opin. Oncol. 2021, 33, 87–94. [Google Scholar] [CrossRef]
  71. Liu, J.F.; Ray-Coquard, I.; Selle, F.; Poveda, A.M.; Cibula, D.; Hirte, H.; Hilpert, F.; Raspagliesi, F.; Gladieff, L.; Harter, P.; et al. Randomized Phase II Trial of Seribantumab in Combination with Paclitaxel in Patients with Advanced Platinum-Resistant or -Refractory Ovarian Cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2016, 34, 4345–4353. [Google Scholar] [CrossRef]
  72. Curley, M.D.; Sabnis, G.J.; Wille, L.; Adiwijaya, B.S.; Garcia, G.; Moyo, V.; Kazi, A.A.; Brodie, A.; MacBeath, G. Seribantumab, an Anti-ERBB3 Antibody, Delays the Onset of Resistance and Restores Sensitivity to Letrozole in an Estrogen Receptor-Positive Breast Cancer Model. Mol. Cancer Ther. 2015, 14, 2642–2652. [Google Scholar] [CrossRef]
  73. Sequist, L.V.; Gray, J.E.; Harb, W.A.; Lopez-Chavez, A.; Doebele, R.C.; Modiano, M.R.; Jackman, D.M.; Baggstrom, M.Q.; Atmaca, A.; Felip, E.; et al. Randomized Phase II Trial of Seribantumab in Combination with Erlotinib in Patients with EGFR Wild-Type Non-Small Cell Lung Cancer. Oncologist 2019, 24, 1095–1102. [Google Scholar] [CrossRef]
  74. Carrizosa, D.R.; Burkard, M.E.; Elamin, Y.Y.; Desai, J.; Gadgeel, S.M.; Lin, J.J.; Waqar, S.N.; Spigel, D.R.; Chae, Y.K.; Cheema, P.K.; et al. CRESTONE: Initial Efficacy and Safety of Seribantumab in Solid Tumors Harboring NRG1 Fusions. J. Clin. Oncol. 2022, 40, 3006. [Google Scholar] [CrossRef]
  75. Patil, T.; Carrizosa, D.R.; Burkard, M.E.; Reckamp, K.L.; Desai, J.; Chae, Y.K.; Liu, S.V.; Konduri, K.; Gadgeel, S.M.; Lin, J.J.; et al. Abstract CT229: CRESTONE: A Phase 2 Study of Seribantumab in Adult Patients with Neuregulin-1 (NRG1) Fusion Positive Locally Advanced or Metastatic Solid Tumors. Cancer Res. 2023, 83, CT229. [Google Scholar] [CrossRef]
  76. Schram, A.M.; Goto, K.; Kim, D.-W.; Martin-Romano, P.; Ou, S.-H.I.; O’Kane, G.M.; O’Reilly, E.M.; Umemoto, K.; Duruisseaux, M.; Neuzillet, C.; et al. Efficacy and Safety of Zenocutuzumab, a HER2 x HER3 Bispecific Antibody, across Advanced NRG1 Fusion (NRG1+) Cancers. J. Clin. Oncol. 2022, 40, 105. [Google Scholar] [CrossRef]
  77. Le, X.; Cornelissen, R.; Garassino, M.; Clarke, J.M.; Tchekmedyian, N.; Goldman, J.W.; Leu, S.-Y.; Bhat, G.; Lebel, F.; Heymach, J.V.; et al. Poziotinib in Non-Small-Cell Lung Cancer Harboring HER2 Exon 20 Insertion Mutations After Prior Therapies: ZENITH20-2 Trial. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2022, 40, 710–718. [Google Scholar] [CrossRef]
  78. Cornelissen, R.; Prelaj, A.; Sun, S.; Baik, C.; Wollner, M.; Haura, E.B.; Mamdani, H.; Riess, J.W.; Cappuzzo, F.; Garassino, M.C.; et al. Poziotinib in Treatment-Naive NSCLC Harboring HER2 Exon 20 Mutations: ZENITH20-4, A Multicenter, Multicohort, Open-Label, Phase 2 Trial (Cohort 4). J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2023, 18, 1031–1041. [Google Scholar] [CrossRef]
  79. Socinski, M.A.; Cornelissen, R.; Garassino, M.C.; Clarke, J.; Tchekmedyian, N.; Molina, J.; Goldman, J.W.; Bhat, G.; Lebel, F.; Le, X. LBA60 ZENITH20, a Multinational, Multi-Cohort Phase II Study of Poziotinib in NSCLC Patients with EGFR or HER2 Exon 20 Insertion Mutations. Ann. Oncol. 2020, 31, S1188. [Google Scholar] [CrossRef]
Figure 1. Schematic of NRG1 gene structure showing position on chromosome 8, composition of exons, and structure of protein isoforms. Ig—immunoglobulin-like domain; S—stalk; EGF-like—EGF-like domain; CT—cytoplasmic tail; CRD—cysteine-rich domain. Black arrows indicate the location of cleavage in secreted types of NRG1.
Figure 1. Schematic of NRG1 gene structure showing position on chromosome 8, composition of exons, and structure of protein isoforms. Ig—immunoglobulin-like domain; S—stalk; EGF-like—EGF-like domain; CT—cytoplasmic tail; CRD—cysteine-rich domain. Black arrows indicate the location of cleavage in secreted types of NRG1.
Cancers 16 02766 g001
Figure 2. The scheme of dimeric ErbB downstream signaling pathways regulated by the NRG1 protein. The NRG1 protein, through different biological cascades, affects the ErbB dimers for protein synthesis, cell survival, cell apoptosis, control of cell cycle and metabolism, as well as cell migration, invasion, or differentiation.
Figure 2. The scheme of dimeric ErbB downstream signaling pathways regulated by the NRG1 protein. The NRG1 protein, through different biological cascades, affects the ErbB dimers for protein synthesis, cell survival, cell apoptosis, control of cell cycle and metabolism, as well as cell migration, invasion, or differentiation.
Cancers 16 02766 g002
Figure 3. The circos plot presents the common fusion partners of NRG1 within chromosome 8.
Figure 3. The circos plot presents the common fusion partners of NRG1 within chromosome 8.
Cancers 16 02766 g003
Figure 4. The circos plot presents the common fusion partners of NRG1 with genes localized within all chromosomes.
Figure 4. The circos plot presents the common fusion partners of NRG1 with genes localized within all chromosomes.
Cancers 16 02766 g004
Table 1. Partner genes, with their chromosomal localization and translocation description, including the NRG1 gene, in different cancer types.
Table 1. Partner genes, with their chromosomal localization and translocation description, including the NRG1 gene, in different cancer types.
Fusion GeneLocalizationAberrationCancer Type
ADAM98p11.22t(8;8)(p12;p11)Breast Cancer
AKAP1315q25.3t(8;15)(p12;q25)
ARHGEF399p13.3t(8;9)(p12;p13)
BABAM2/BRE2p23.2t(8;2)(p12;p23)
CD912p13.31t(8;12)(p12;p13)
COX10-AS117p12t(8;17)(p12;p12)
DDHD28p11.23t(8;8)(p12;p11)
FAM91A18q24.13t(8;8)(p12;q24)
FOXA114q21.1t(8;14)(p12;q21)
FUT108p12t(8;8)(p12;p12)
TENM411q14.1t(8;11)(p12;q14)
ZNF7048q21.13t(8;8)(p12;q21)
ATP1B11q24.2t(8;1)(p12;q24)Breast Cancer/Cholangiocarcinoma/Pancreatic Ductal Adenocarcinoma
ERO1L14q22.1t(8;14)(p12;q22)
IKBKB8p11.21t(8;8)(p12;p11)Colorectal Cancer
KCTD9 8p21.2t(8;8)(p12;q21)
POMK8p11.21t(8;8)(p12;p11)
TNFRSF10B8p21.3t(8;8)(p12;p21)
ZCCHC79p13.2t(8;9)(p12;p13)
BIN38p21.3t(8;8)(p12;p21)Esophageal Carcinoma
CCAR28p21.3t(8;8)(p12;p21)
NOTCH21p12t(8;1)(p12;p12)Gallbladder Cancer
PDE7A8q13.1t(8;8)(p12;q13)Head and Neck Squamous Cell Carcinoma
THBS115q14t(8;15)(p12;q14)
PCM18p22t(8;8)(p12;p22)Kidney Renal Clear Cell Carcinoma
CD745q33.1t(8;5)(p12;q33)Lung Adenocarcinoma/Pancreatic Adenocarcinoma
CADM111q23.3t(8;11)(p12;q23)Lung Cancer
DIP2B12q13.12t(8;12)(p12;q13)
DPYSL28p21.2t(8;8)(p12;p21)
F11R1q23.3t(8;1)(p12;q23)
FGFR18p11.23t(8;8)(p12;q11)
FLYWCH116p13.3t(8;16)(p12;p13)
ITGB110p11.22t(8;10)(p12;p11)
KIF13B8p12t(8;8)(p12;p12)
KRAS12p12.2t(8;12)(p12;p12)
MDK11p11.2t(8;11)(p12;p11)
MRPL138q24.12t(8;8)(p12;q24)
NPTN15q24.1t(8;15)(p12;q24)
PARP85q11.1t(8;5)(p12;q11)
PLCG216q23.3t(8;16)(p12;q23)
RALGAPA114q13.2t(8;14)(p12;q13)
SDC420q13.12t(8;20)(p12;q13)
SLC3A211q12.3t(8;11)(p12;q12)
SMAD418q21.2t(8;18)(p12;q21)
THAP722q11.21t(8;22)(p12;q11)
TNC9q33.1t(8;9)(p12;q33)
WRN8p12t(8;8)(p12;p12)Lung Cancer/Breast Cancer
RBPMS8p12t(8;8)(p12;p12)Lung Cancer/Renal Cell Carcinoma
HMBOX18p21.1-p12t(8;8)(p12;p21)Neuroendocrine Tumor of the Nasopharynx/Spindle Cell Sarcoma
CLU8p21.1t(8;8)(p12;p21)Ovarian Cancer
RAB3IL111q12.2-q12.3t(8;11)(p12;q12)
SETD421q22.12t(8;21)(p12;q22)
TSHZ220q13.2t(8;20)(p12;q13)
ZMYM213q12.11t(8;13)(p12;q11)
APP21q21.3t(8;21)(p12;q21)Pancreatic Adenocarcinoma
CDH116q22.1t(8;16)(p12;q22)
CDH65p13.3t(8;5)(p12;p13)
CDK110q21.2t(8;10)(p12;q21)
ROCK118q11.1t(8;18)(p12;q11)
SARAF8p12t(8;8)(p12;p12)
UNC5D8p12t(8;8)(p12;p12)
VTCN11p13.1-p12t(8;1)(p12;p13)
STMN28q21.13t(8;8)(p12;q21)Prostate Cancer
WHSC1L18p11.23t(8;8)(p12;p11)Sarcoma
MTUS18p22t(8;8)(p12;p22)Spindle Cell Sarcoma
PPHLN112q12t(8;12)(p12;q12)
GDF1519p13.11t(8;19)(p12;p13)Urothelial Bladder Cancer
PMEPA120q13.31t(8;20)(p12;q13)Uterine Carcinosarcoma
Table 2. A summary of the effect of different drugs on the prognosis of lung cancer patients.
Table 2. A summary of the effect of different drugs on the prognosis of lung cancer patients.
DrugStudied MaterialEffectReference
AfatinibNSCLC patients10–12 months of durable responseGay et al. [8]
5–27 months of partial response
4 months of stable disease
Cadranel et al. [54]
7 months of partial responseWu et al. [55]
48.3% of the overall response rate
6.8 months median duration of response
6.1 months median progression-free survival
Liu et al. [56]
Tarloxotinibpatient-derived cell lines, murine xenograft modelsinhibition of tumor growth
cancer regression
Bhandari et al. [57]
Estrada-Bernal et al. [58]
Seribantumabpatient-derived cell lines, patient-derived xenograft modelsreduction of proliferation
induction of apoptosis
Odintsov et al. [59]
patient-derived xenograft mouse model MDA-MB-175-VII cell linedurable tumor regression
anti-proliferative activity
Drilon et al. [25]
Table 3. A summary of clinical trials dedicated to patients with solid tumors (including NSCLC) harboring NRG1 fusions. Data were collected from the ClinalTrials.gov database (http://clinicaltrials.gov/ (accessed on 30 July 2024)).
Table 3. A summary of clinical trials dedicated to patients with solid tumors (including NSCLC) harboring NRG1 fusions. Data were collected from the ClinalTrials.gov database (http://clinicaltrials.gov/ (accessed on 30 July 2024)).
Clinical Trial ID (Duration)
Status
Tested Drug (Phase)Genetic EligibilityConditions (Cohort)Primary Measured Outcomes
NCT05919537
(09.2023–03.2031)
Recruiting
HMBD-001 with/without chemotherapy
(Phase I)
NRG1 fusions
Extracellular domain HER3 mutations
Advanced solid tumors
(68)
1. Adverse events
2. Incidence and nature of dose-limiting toxicities (DLTs)
3. ORR
NCT02912949
(01.2015–12.2026)
Recruiting
Zenocutuzumab (MCLA-128)
(Phase 2)
NRG1 fusionsSolid tumors
(250)
1. ORR
2. Duration of response
NCT04383210
(09.2020–03-2025)
Active, not-recruiting
Seribantumab
(Phase 2)
NRG1 fusionsLocally advanced or metastatic solid tumors
(75)
ORR
NCT04750824
(10.2020–12.2021)
Completed
Afatinib
(Observational)
NRG1 fusionsSolid tumors
(110)
ORR
NCT05057013
(11.2021–09.2026)
Recruiting
HMBD-001
(Phase 1)
NRG1 fusions
HER3 expression
Solid tumors
(135)
1. Recommended dose
2. Adverse events
3. ORR
NCT03805841
(03.2019–04.2021)
Terminated
Tarloxotinib
(Phase 2)
NRG1 fusions
ERBB family fusions
EGFR Exon 20 Insertion
HER2-activating mutations
NSCLC or advanced solid tumors
(41)
ORR
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kucharczyk, T.; Nicoś, M.; Kucharczyk, M.; Kalinka, E. NRG1 Gene Fusions—What Promise Remains Behind These Rare Genetic Alterations? A Comprehensive Review of Biology, Diagnostic Approaches, and Clinical Implications. Cancers 2024, 16, 2766. https://doi.org/10.3390/cancers16152766

AMA Style

Kucharczyk T, Nicoś M, Kucharczyk M, Kalinka E. NRG1 Gene Fusions—What Promise Remains Behind These Rare Genetic Alterations? A Comprehensive Review of Biology, Diagnostic Approaches, and Clinical Implications. Cancers. 2024; 16(15):2766. https://doi.org/10.3390/cancers16152766

Chicago/Turabian Style

Kucharczyk, Tomasz, Marcin Nicoś, Marek Kucharczyk, and Ewa Kalinka. 2024. "NRG1 Gene Fusions—What Promise Remains Behind These Rare Genetic Alterations? A Comprehensive Review of Biology, Diagnostic Approaches, and Clinical Implications" Cancers 16, no. 15: 2766. https://doi.org/10.3390/cancers16152766

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop