her3 expression
Recently Published Documents


TOTAL DOCUMENTS

68
(FIVE YEARS 22)

H-INDEX

11
(FIVE YEARS 4)

2021 ◽  
Author(s):  
Mari Iida ◽  
Nellie McDaniel ◽  
Kourtney Kostecki ◽  
Noah Welke ◽  
Carlene Kranjac ◽  
...  

Abstract Background The receptor tyrosine kinase (RTK) epidermal growth factor receptor (EGFR) is overexpressed and an important therapeutic target in Head and Neck cancer (HNC). Cetuximab is currently the only EGFR-targeting agent approved by the FDA for treatment of HNC; however, intrinsic and acquired resistance to cetuximab is a major problem in the clinic. Our lab previously reported that AXL leads to cetuximab resistance via activation of HER3. Methods In this study, we investigate the connection between AXL, HER3, and neuregulin1 (NRG1) gene expression with a focus on understanding how their interdependent signaling promotes resistance to cetuximab in head and neck cancer. Plasmid or siRNA transfections, cell proliferation assays, and clonogenic assays were conducted to test cetuximab sensitivity. Quantitative PCR and immunoblot analysis were used to analyze gene expression levels. Seven HNC patient-derived xenografts (PDXs) were evaluated for protein expression levels. Results We found that HER3 expression was necessary but not sufficient for cetuximab resistance without AXL expression. Our results demonstrated that addition of the HER3 ligand NRG1 to cetuximab-sensitive HNC cells leads to cetuximab resistance. Further, AXL-overexpressing cells regulate NRG1 at the level of transcription, thereby promoting cetuximab resistance. Immunoblot analysis revealed that NRG1 expression was relatively high in cetuximab-resistant HNC PDXs compared to cetuximab-sensitive HNC PDXs. Finally, genetic inhibition of NRG1 resensitized AXL-overexpressing cells to cetuximab. Conclusions The results of this study indicate that AXL may signal through HER3 via NRG1 to promote cetuximab resistance and that targeting of NRG1 could have significant clinical implications for HNC therapeutic approaches.


2021 ◽  
Vol 9 (11) ◽  
pp. e003616
Author(s):  
Nadine Aschmoneit ◽  
Lennart Kühl ◽  
Oliver Seifert ◽  
Roland E Kontermann

BackgroundBispecific T-cell engagers are an established therapeutic strategy for the treatment of hematologic malignancies but face several challenges when it comes to their application for the treatment of solid tumors, including on-target off-tumor adverse events. Employing an avidity-mediated specificity gain by introducing an additional binding moiety for the tumor-associated antigen can be achieved using formats with a 2+1 stoichiometry.MethodsBesides biochemical characterization and validation of target cell binding to cancer cells with different HER3 expression, we used in vitro co-culture assays with human peripheral blood mononuclear cells (PBMCs) and HER3-expressing target cells to determine T-cell activation, T-cell proliferation and PBMC-mediated cancer cell lysis of HER3-positive cell lines by the trivalent, bispecific antibodies.ResultsIn this study, we developed trivalent, bispecific antibodies comprising a silenced Fc region for T-cell retargeting to HER3-expressing tumor cells, combining a bivalent single-chain diabody (scDb) fused to a first heterodimerizing Fc chain with either an Fab or scFv fused to a second heterodimerizing Fc chain. All these HER3-targeting T-cell engagers comprising two binding sites for HER3 and one binding site for CD3 mediated target cell killing. However, format and orientation of binding sites influenced efficacy of target cell binding, target cell-dependent T-cell activation and T-cell-mediated target cell killing. Beneficial effects were seen when the CD3 binding site was located in the scDb moiety. These molecules showed efficient killing of medium HER3-expressing cancer cells with very low induction of cytokine release, while sparing target cells with low or undetectable HER3 expression.ConclusionOur study demonstrates that these trivalent, bispecific antibodies represent formats with superior interdomain spacing resulting in efficient target cell killing and a potential advantageous safety profile due to very low cytokine release.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Giovanni Almadori ◽  
Antonella Coli ◽  
Eugenio De Corso ◽  
Dario Antonio Mele ◽  
Stefano Settimi ◽  
...  

Abstract Background Compared to the other members of human epidermal growth factor family receptors (HER), the role of HER3 has not been well defined in laryngeal cancer. The predictive and prognostic role of HER3 has been the focus of clinical attention but the research findings are contradictory, especially in laryngeal squamous cell carcinoma (LSCC). The variable localization of HER3 within cancer cells and the role of HER3 in primary and acquired resistance to HER1-targeted therapies remain unclear. Methods We performed a retrospective analysis of two cohorts of 66 homogeneous consecutive untreated primary advanced LSCC patients, in which co-expression of HER1, HER2 and HER3 receptors was investigated by semi-quantitative immunohistochemistry. The association of their pattern of expression with survival was evaluated by Kaplan–Meier and Cox’s proportional hazard analyses. Multivariable Cox proportional hazards models were developed to predict median 2- and 3-year RFS and 2.5- and 5-year OS. The Akaike information criterion technique and backwards stepwise procedure were used for model selections. The performance of the final Cox models was assessed with respect to calibration and discrimination. Results Immunohistochemical labeling for HER1 and HER2 was localized both in the cell membrane and in the cytoplasm, while HER3 labeling was observed both in the cell cytoplasm and in the nucleus. HER3 expression was inversely correlated with HER1 positivity. The expression patterns of HERs were associated with tumor differentiation. In both cohorts of patients, HER1 expression was associated with reduced relapse-free (RFS) and overall survival (OS). In HER1 positive tumors, the co-expression with nuclear HER3 was associated with better RFS and OS, compared with HER3 negative tumors or tumors expressing HER3 at cytoplasmic level. HER3 expressing tumors had a higher Geminin/MCM7 ratio than HER3 negative ones, regardless of HER1 co-expression. Multivariable analyses identified age at diagnosis, tumor site, HER1, HER3 and age at diagnosis, tumor stage, HER1, HER3, as covariates significantly associated with RFS and OS, respectively. Bootstrapping verified the good fitness of these models for predicting survivals and the optimism-corrected C-indices were 0.76 and 0.77 for RFS and OS, respectively. Conclusions Nuclear HER3 expression was strongly associated with favourable prognosis and allows to improve the prognostic stratification of patients with HER1 positive advanced LSCC carcinoma.


Cancers ◽  
2021 ◽  
Vol 13 (19) ◽  
pp. 4791
Author(s):  
Sara S. Rinne ◽  
Charles Dahlsson Leitao ◽  
Ayman Abouzayed ◽  
Anzhelika Vorobyeva ◽  
Vladimir Tolmachev ◽  
...  

HER3 (human epidermal growth factor receptor type 3) is a challenging target for diagnostic radionuclide molecular imaging due to the relatively modest overexpression in tumors and substantial expression in healthy organs. In this study, we compared four HER3-targeting PET tracers based on different types of targeting molecules in a preclinical model: the 89Zr-labeled therapeutic antibody seribantumab, a seribantumab-derived F(ab)2-fragment labeled with 89Zr and 68Ga, and the 68Ga-labeled affibody molecule [68Ga]Ga-ZHER3. The novel conjugates were radiolabeled and characterized in vitro using HER3-expressing BxPC-3 and DU145 human cancer cells. Biodistribution was studied using Balb/c nu/nu mice bearing BxPC-3 xenografts. HER3-negative RAMOS xenografts were used to demonstrate binding specificity in vivo. Autoradiography was conducted on the excised tumors. nanoPET/CT imaging was performed. New conjugates specifically bound to HER3 in vitro and in vivo. [68Ga]Ga-DFO-seribantumab-F(ab’)2 was considered unsuitable for imaging due to the low stability and high uptake in normal organs. The highest tumor-to-non-tumor contrast with [89Zr]Zr-DFO-seribantumab and [89Zr]Zr-DFO-seribantumab-F(ab’)2 was achieved at 96 h and 48 h pi, respectively. Despite lower tumor uptake, [68Ga]Ga-ZHER3 provided the best imaging contrast due to the fastest clearance from blood and normal organs. The results of our study suggest that affibody-based tracers are more suitable for PET imaging of HER3 expression than antibody- and antibody-fragment-based tracers.


Oncogenesis ◽  
2021 ◽  
Vol 10 (8) ◽  
Author(s):  
Yi-Ping Wang ◽  
I-Ju Liu ◽  
Kai-Chi Chen ◽  
Han-Chung Wu

AbstractEpidermal growth factor receptor (EGFR) remains the sole druggable molecular target other than the PD1/PD-L1 pathway with meaningful clinical benefit in squamous cell carcinoma of head and neck (SCCHN). Human epidermal growth factor receptor 3 (HER3) confers the resistance to EGFR-targeted treatment in SCCHN. Thus, it is essential to determine the distribution and regulatory mechanisms of HER3 in SCCHN. We explored the prevalence of HER3 expression and its distribution within SCCHN by immunohistochemical staining and clinicopathological correlations were analyzed. The regulatory mechanism of HER3 expression was then dissected in vitro, using RT-PCR, Western blotting, and immunoprecipitation in a set of SCCHN cell lines. Subsequent in vivo validation in the murine model was also performed. We found that concomitant high expression of HER3 and its ligand NRG1 in SCCHN is associated with the increased presence of regional lymphatic metastasis and the majority of HER3 is located on the differentiated tumor cells. Further investigation revealed that HER3 is under positive control of NOTCH1 through transcriptional activation and inhibition of protein degradation through the polyubiquitination machinery via AKT pathway and USP8 deubiquitinating enzyme. In addition, loss of function of NOTCH1 suppresses HER3 expression through increased phosphorylation of serine 473 of AKT in SCCHN cells, and promotes the aggressiveness of the tumor cells. These data indicated that the level of HER3 is regulated by NOTCH1 in SCCHN both transcriptionally and post-translationally, and NOTCH1 is in a higher hierarchy in the regulatory system of the AKT pathway. Since NOTCH1 is inactivated in approximately 10% of SCCHN cases and this aberration strongly impacts the AKT pathway and diminishes HER3, exclusion of patients with NOTCH1-inactivated SCCHN may be beneficial for future clinical trials of HER3-targeting antibodies.


2021 ◽  
Vol 27 (14) ◽  
pp. 4129-4129
Author(s):  
Toni M. Brand ◽  
Stefan Hartmann ◽  
Neil E. Bhola ◽  
Noah D. Peyser ◽  
Hua Li ◽  
...  

2021 ◽  
Vol 32 ◽  
pp. S296
Author(s):  
Kimio Yonesaka ◽  
Junko Tanizaki ◽  
Osamu Maenishi ◽  
Hisato Kawakami ◽  
Kaoru Tanaka ◽  
...  

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS9139-TPS9139
Author(s):  
Pasi A. Janne ◽  
Melissa L. Johnson ◽  
Yasushi Goto ◽  
James Chih-Hsin Yang ◽  
Michele Vigliotti ◽  
...  

TPS9139 Background: Few treatment options have demonstrated therapeutic benefit in epidermal growth factor receptor–mutated ( EGFRm) non–small cell lung cancer (NSCLC) that has progressed after treatment with EGFR tyrosine kinase inhibitors (TKIs) and platinum-based chemotherapy. HER3, a member of the human epidermal growth factor family, is detectable in most EGFRm NSCLC, and its expression has been linked to worse clinical outcomes. There are no approved HER3 directed therapies for the treatment of NSCLC. HER3-DXd is a novel, potentially first-in-class HER3 directed antibody drug conjugate that has demonstrated preliminary evidence of safety and antitumor activity in patients (pts) with EGFRm TKI–resistant NSCLC in an ongoing Phase 1 study, providing proof of concept of HER3-DXd. The Phase 2 study (HERTHENA-Lung01) is further evaluating HER3-DXd in pts with previously treated metastatic or locally advanced EGFRm NSCLC. Methods: This randomized, open-label Phase 2 study will enroll up to 420 pts at approximately 135 study sites in North America, Europe and the Asia-Pacific region. Eligible pts will have metastatic or locally advanced NSCLC with an activating EGFR mutation (exon 19 deletion or L858R), progression during or after systemic treatment with ≥1 EGFR TKI and ≥1 platinum-based chemotherapy regimen, and ≥1 measurable lesion confirmed by blinded independent central review (BICR) per RECIST v1.1. Pts with an EGFR T790M mutation must have received and progressed on prior osimertinib. Pts with stable brain metastases are eligible. Exclusion criteria include evidence of previous small cell or combined small cell/non–small cell histology or any history of interstitial lung disease. Tumor tissue will be assessed retrospectively for HER3 expression and molecular mechanisms of TKI resistance. HER3 expression will not be used to select pts for enrollment. Pts will be randomized 1:1 to receive 1 of 2 HER3-DXd Q3W dose regimens that will be independently evaluated: a 5.6 mg/kg fixed-dose regimen (Arm 1) or an up-titration dose regimen (Arm 2: Cycle 1, 3.2 mg/kg; Cycle 2, 4.8 mg/kg; Cycle 3 and beyond, 6.4 mg/kg). After review of data from an ongoing Phase 1 study with similar patients treated with either of these dose regimens, a decision could be made to continue enrollment into 1 or both arms. The primary objective is to evaluate the efficacy of HER3-DXd as measured by objective response rate (ORR) by BICR. Secondary objectives are to evaluate the efficacy and safety/tolerability of HER3-DXd and to assess the relationship between efficacy and HER3 expression. Secondary endpoints include duration of response, progression-free survival, ORR by investigator, disease control rate, time to response, best percentage change in the sum of diameters of measurable tumors, and overall survival. The study is enrolling and is planned to finish in 2023. Clinical trial information: NCT04619004.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Avisek Majumder ◽  
Manbir Sandhu ◽  
Debarko Banerji ◽  
Veronica Steri ◽  
Adam Olshen ◽  
...  

AbstractHER2 and HER3 play key driving functions in the pathophysiology of HER2-amplified breast cancers, but this function is less well characterized in other cancers driven by HER2 amplification. This study aimed to explore the role of HER2 and HER3 signaling in other types of HER2-amplified cancer. The expression and signaling activity of HER2, HER3, and downstream pathway proteins were studied in cell panels representing HER2-amplified cancers of the breast, bladder, colon and rectal, stomach, esophagus, lung, tongue, and endometrium along with controls lacking HER2 amplification. We report that HER2-amplified cancers are addicted to HER2 across different cancer types and the depth of addiction is best linked with the expression level of HER2, but not with HER3 expression. We report that the expression and constitutive phosphorylation of HER3 are ubiquitous in HER2-amplified breast cancer cell lines, but much more variable in HER2-amplified cancer cells from other tissues. We observed the lapatinib-induced compensatory upregulation of HER3 signaling in many types of HER2-amplified cancers, although with much variability. We find that HER3 expression is essential for in vivo tumorigenic growth in some HER2-amplified tumors but not others. Importantly HER3 expression level does not correlate well with its functional importance. More biomarkers will be needed to guide the optimal use of HER3 inhibitors in HER2-amplified cancers from non-breast origin. Unlike oncogenes activated through mutational events, the activation of HER2 through overexpression represents a gradient of activities and depth of addiction and the response to inhibitors follows a similar gradient.


2021 ◽  
pp. LMT48
Author(s):  
Thubeena Manickavasagar ◽  
Wei Yuan ◽  
Suzanne Carreira ◽  
Bora Gurel ◽  
Susana Miranda ◽  
...  

Aim: We explore HER3 expression in lung adenocarcinoma (adeno-NSCLC) and identify potential mechanisms of HER3 expression. Materials & methods: Tumor samples from 45 patients with adeno-NSCLC were analyzed. HER3 and HER2 expression were identified using immunohistochemistry and bioinformatic interrogation of The Cancer Genome Atlas (TCGA). Results: HER3 was highly expressed in 42.2% of cases. ERBB3 copy number did not account for HER3 overexpression. Bioinformatic analysis of TCGA demonstrated that MEK activity score (a surrogate of functional signaling) did not correlate with HER3 ligands. ERBB3 RNA expression levels were significantly correlated with MEK activity after adjusting for EGFR expression. Conclusion: HER3 expression is common and is a potential therapeutic target by virtue of frequent overexpression and functional downstream signaling.


Sign in / Sign up

Export Citation Format

Share Document