Mutations inducing resistance to anti-epidermal growth factor receptor (EGFR) therapy may have a clinical impact even if present in minor cell clones which could expand during treatment. We tested this hypothesis in lung cancer patients treated with tyrosine kinase inhibitors (TKIs). Eighty-three patients with lung adenocarcinoma treated with erlotinib or gefitinib were included in this study. The mutational status of KRAS and EGFR was investigated by direct sequencing (DS). KRAS mutations were also assessed by mutant-enriched sequencing (ME-sequencing). DS detected KRAS mutations in 16 (19%) of 83 tumors; ME-sequencing identified all the mutations detected by DS but also mutations in minor clones of 14 additional tumors, for a total of 30 (36%) of 83. KRAS mutations assessed by DS and ME-sequencing significantly correlated with resistance to TKIs (P = .04 and P = .004, respectively) and significantly affected progression-free survival (PFS) and overall survival (OS). However, the predictive power of mutations assessed by ME-sequencing was higher than that obtained by DS (hazard ratio [HR] = 2.82, P = .0001 vs HR = 1.98, P = .04, respectively, for OS; HR = 2.52, P = .0005 vs HR = 2.21, P = .007, respectively, for PFS). Survival outcome of patients harboring KRAS mutations in minor clones, detected only by ME-sequencing, did not differ from that of patients with KRAS mutations detected by DS. Only KRAS mutations assessed by ME-sequencing remained an independent predictive factor at multivariate analysis. KRAS mutations in minor clones have an important impact on response and survival of patients with lung adenocarcinoma treated with EGFR-TKI. The use of sensitive detection methods could allow to more effectively identify treatment-resistant patients.
Over the last ten years, several new and therapeutically relevant cancer drugs targeting tyrosine kinases signaling pathways have been developed. Tyrosine kinase inhibitors (TKIs) are a pharmaceutical class of small molecules, orally available, well-tolerated, worldwide approved drugs for the treatment of several neoplasms, including lung, breast, kidney and pancreatic cancer as well as gastro-intestinal stromal tumors and chronic myeloid leukemia. This comprehensive review focuses on the most relevant members of the first and the second generation TKIs designed to interact with receptor and nonreceptor TKs. Attention is mainly focused on molecular mechanisms in in vitro and in vivo models related to the clinical activity of the drugs and to the development of resistance to treatment, still the major challenge in cancer research and care.
We aimed to overcome intratumoral heterogeneity in clear cell renal cell carcinoma (clearRCC). One hundred cases of clearRCC were sampled. First, usual standard sampling was applied (1 block/cm of tumor); second, the whole tumor was sampled, and 0.6 mm cores were taken from each block to construct a tissue microarray; third, the residual tissue, mapped by taking pieces 0.5 × 0.5 cm, reconstructed the entire tumor mass. Precisely, six randomly derived pieces of tissues were placed in each cassette, with the number of cassettes being based on the diameter of the tumor (called multisite 3D fusion). Angiogenic and immune markers were tested. Routine 5231 tissue blocks were obtained. Multisite 3D fusion sections showed pattern A, homogeneous high vascular density (10%), pattern B, homogeneous low vascular density (8%) and pattern C, heterogeneous angiogenic signatures (82%). PD-L1 expression was seen as diffuse (7%), low (33%) and absent (60%). Tumor-infiltrating CD8 scored high in 25% (pattern hot), low in 65% (pattern weak) and zero in 10% of cases (pattern desert). Grading was upgraded in 26% of cases (G3–G4), necrosis and sarcomatoid/rhabdoid characters were observed in, respectively, 11 and 7% of cases after 3D fusion (p = 0.03). CD8 and PD-L1 immune expressions were higher in the undifferentiated G4/rhabdoid/sarcomatoid clearRCC subtypes (p = 0.03). Again, 22% of cases were set to intermediate to high risk of clinical recurrence due to new morphological findings of all aggressive G4, sarcomatoid/rhabdoid features by using 3D fusion compared to standard methods (p = 0.04). In conclusion, we propose an easy-to-apply multisite 3D fusion sampling that negates bias due to tumor heterogeneity.
Background: HER2 mutations are oncogenic in hormone receptor positive (HR+) metastatic breast cancer (MBC), and may confer resistance to prior endocrine therapy but retain sensitivity to neratinib. Neratinib is an oral, irreversible, pan-HER tyrosine kinase inhibitor with clinical activity either as a single agent or in combination with fulvestrant in HER2-mutated, HER2-non-amplified MBC. Genomic analyses suggest that acquired resistance to neratinib can occur via additional HER2 alterations, which may alter HER2-pathway signaling. We investigated whether dual HER2-targeted therapy could improve clinical benefit in a cohort of patients with HER2-mutant, HR+ MBC treated with neratinib + trastuzumab + fulvestrant (N+T+F) from SUMMIT - a phase 2 basket trial (NCT01953926). Methods: Patients with HR+ MBC with known or suspected pathogenic HER2 mutation(s) identified by genomic sequencing were eligible to receive N+T+F (oral neratinib 240 mg/day, i.v. trastuzumab 8 mg/kg initially followed by 6 mg/kg every 3 weeks, and i.m. fulvestrant 500 mg on days 1&15 of month 1, then on day 1 every 4 weeks). Loperamide prophylaxis was mandatory during the first 2 treatment cycles. There was no restriction on the number of prior lines of systemic treatment for MBC. Efficacy endpoints: confirmed objective response rate and clinical benefit rate (RECIST v1.1); duration of response; progression-free survival. Results: As of 22-May-2020, 46 patients were enrolled in the N+T+F cohort and received at least 1 dose of study medication (safety population). 14 unique HER2 allelic variants were identified: 8 kinase domain missense; 1 extracellular domain missense; 2 transmembrane domain missense; 2 exon-20 insertion; 1 exon-19 deletion. The most common HER2 mutant variant was L755S (n=15, 33%) Median number of prior systemic regimens for metastatic disease was 4 (range 0-10); 34 (74%) patients had received prior fulvestrant, and 31 (67%) patients had received prior cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitor therapy. 16 (35%) patients had ductal histology, 29 (63%) had lobular carcinoma, and 1 (2%) had mixed ductal and lobular carcinoma. At this time, 30/46 patients had RECIST measurable disease and are efficacy evaluable (ongoing patients who did not have the opportunity for their first post-baseline tumor assessment were excluded); clinical activity - see Table. Diarrhea was the most commonly reported adverse event (80% any grade) with 15 (33%) patients reporting grade 3 diarrhea (no grade 4 diarrhea). 10 patients (22%) had a neratinib dose reduction due to diarrhea but no patients discontinued treatment due to diarrhea. Conclusions: The combination of N+T+F demonstrated encouraging clinical activity in heavily pre-treated HER2-mutant, HR+, HER2-non-amplified MBC, including patients who had previously received either fulvestrant and/or CDK4/6 inhibitor-based therapies. While the rate of grade 3 diarrhea was higher than that observed with single-agent neratinib in SUMMIT, this was manageable through loperamide prophylaxis, and no patients discontinued study treatment due to diarrhea. SUMMIT has recently been amended to evaluate N+T+F, T+F and F (1:1:1 randomization) and continues to enroll patients. RECIST measurable and efficacy evaluable patients (n=30)Confirmed objective response,a n (%)12 (40)CR0PR12ORR, % (95% CI)40 (23-59)Best overall response, n (%)18 (60)CR0PR18Best overall response rate, % (95% CI)60 (41-77)Medianb DOR, months (95% CI)8.4 (4.1-NE)Clinical benefit,c n (%)14 (47)CR or PR12SD ≥24 weeks2CBR, % (95% CI)47 (28-66)Medianb PFS, months (95% CI)8.3 (4.2-12.5)aORR is defined as either a CR or a PR that is confirmed no less than 4 weeks after the criteria for response are initially met; bKaplan-Meier analysis; cCBR is defined as confirmed CR or PR or SD for ≥24 weeks; CR, complete response; CBR, clinical benefit rate; DOR, duration of response; NE, not estimable; ORR, objective response rate; PFS, progression-free survival; PR, partial response; SD, stable disease. Citation Format: Komal Jhaveri, Cristina Saura, Angel Guerrero-Zotano, Iben Spanggaard, François-Clement Bidard, Jonathan W Goldman, José A García-Sáenz, Andrés Cervantes, Valentina Boni, John Crown, Adam Brufsky, Sherene Loi, Barbara Haley, Ingrid A Mayer, Stephen Chia, Janice Lu, James Waisman, Noa Efrat Ben-Baruch, Mark E Burkard, Jennifer M Suga, Lucía González-Cortijo, Bruno Perrucci, Feng Xu, Sofia Wong, Jie Zhang, Lisa D Eli, Alshad S Lalani, Hans Wildiers. Latest findings from the breast cancer cohort in SUMMIT - a phase 2 ‘basket’ trial of neratinib + trastuzumab + fulvestrant for HER2-mutant, hormone receptor-positive, metastatic breast cancer [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PD1-05.
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.
customersupport@researchsolutions.com
10624 S. Eastern Ave., Ste. A-614
Henderson, NV 89052, USA
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.
Copyright © 2024 scite LLC. All rights reserved.
Made with 💙 for researchers
Part of the Research Solutions Family.