GLORIA

GEOMAR Library Ocean Research Information Access

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
Filter
  • American Association for Cancer Research (AACR)  (19)
Materialart
Verlag/Herausgeber
  • American Association for Cancer Research (AACR)  (19)
Sprache
Erscheinungszeitraum
Fachgebiete(RVK)
  • 1
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 38-38
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 38-38
    Kurzfassung: Purpose: Translational research in non-small cell lung cancer (NSCLC) has been largely hampered by relatively low success rates for establishing patient-derived cell model or patient-derived xenograft model. Recently, NSCLC patient-derived organoid model (PDO) has been introduced, showing a high take rate and stable growth in long-term in vitro cultures. Translational relevance of NSCLC PDOs remains unclear. In this study, we evaluated whether NSCLC PDOs can predict anti-cancer treatment responses and provide therapeutic strategies to overcome drug resistance. Experimental design: Nineteen malignant effusions and 13 biopsied/surgical specimens were obtained from 31 patients with lung adenocarcinoma. Eleven patients (35%) had no prior therapy, 7 (23%) chemotherapy or radiotherapy, 6 (19%) EGFR-tyrosine kinase inhibitors (TKI), 4 (13%) ALK/ROS1-TKIs, and 3 (10%) immunotherapy. Samples were processed and cultured as reported previously. In brief, specimens were reviewed by pathologists to confirm malignancy. Then, samples were cultured in Advanced DMEM/F12 containing 10% R-spondin 1 conditioned medium, 25 ng/mL FGF7, 100 ng/mL FGF10, 100 ng/mL noggin, 500 nM A83-01, 500 nM SB202190, 1X B27, 1.25 mM N-acetylcysteine, 5 mM nicotinamide, 1X glutamax, 10 mM HEPES, and 1X primocin. For efficient establishment, we stained organoids with H & E at early passages to confirm histological features of NSCLC. Cell viability assay was performed using CellTiter-Glo 3D. PDOs ( & lt;P10) were analyzed by whole-exome sequencing and RNA-seq and cryopreserved to establish a biobank. Results: A total of 22 PDOs derived from NSCLC patients was established. Take rates from malignant effusions and biopsied/surgical specimens were 89% and 38%. Of these established PDOs, 9 were wild-type, 9 EGFR-mutant, 3 ROS1-rearranged, and 1 ALK-rearranged. Genomic and transcriptomic profile of established PDOs were concordant to those of matching parental tumors. Of note, we established clinically important models progressing to targeted therapy or immunotherapy as follows: osimertinib-resistant EGFR mutant PDOs (n=4), repotrectinib-resistant ROS1-rearranged PDO (n=1), and pembrolizumab-resistant PDO (n=1). For example, YUO-2 was established from progressing pleural effusion after 5 months of osimertinib and had lost EGFR T790M but maintained EGFR exon 19 deletion. YUO-2 was resistant to gefitinib, afatinib, and osimertinib with IC50 values of 27300 nM, 9296 nM, and 5691 nM, respectively, implicating clinical relevance of NSCLC PDO models. TKI-resistant PDOs were screened with combinations of TKI targeting the driver mutation of the established models and each drug from the FDA-approved drug library. Conclusions: NSCLC PDO models could recapitulate the characteristics of corresponding NSCLC tumors and clinical response. PDO-based co-clinical trial will accelerate translational research in NSCLC. Further results will be presented at 2019 AACR meeting. Citation Format: Seok-Young Kim, Dong Hwi Kim, Hyeong-Seok Joo, Mi Ran Yun, Ji Yeon Lee, Sang Min Kim, Hyunki Kim, Min Hee Hong, Hye Ryun Kim, Byoung Chul Cho. NSCLC patient-derived organoids to guide personalized therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 38.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2019
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 10, No. 8 ( 2020-08-01), p. 1194-1209
    Kurzfassung: EGFR exon 20 insertion driver mutations (Exon20ins) in non–small cell lung cancer (NSCLC) are insensitive to EGFR tyrosine kinase inhibitors (TKI). Amivantamab (JNJ-61186372), a bispecific antibody targeting EGFR–MET, has shown preclinical activity in TKI-sensitive EGFR-mutated NSCLC models and in an ongoing first-in-human study in patients with advanced NSCLC. However, the activity of amivantamab in Exon20ins-driven tumors has not yet been described. Ba/F3 cells and patient-derived cells/organoids/xenograft models harboring diverse Exon20ins were used to characterize the antitumor mechanism of amivantamab. Amivantamab inhibited proliferation by effectively downmodulating EGFR–MET levels and inducing immune-directed antitumor activity with increased IFNγ secretion in various models. Importantly, in vivo efficacy of amivantamab was superior to cetuximab or poziotinib, an experimental Exon20ins-targeted TKI. Amivantamab produced robust tumor responses in two Exon20ins patients, highlighting the important translational nature of this preclinical work. These findings provide mechanistic insight into the activity of amivantamab and support its continued clinical development in Exon20ins patients, an area of high unmet medical need. Significance: Currently, there are no approved targeted therapies for EGFR Exon20ins–driven NSCLC. Preclinical data shown here, together with promising clinical activity in an ongoing phase I study, strongly support further clinical investigation of amivantamab in EGFR Exon20ins–driven NSCLC. This article is highlighted in the In This Issue feature, p. 1079
    Materialart: Online-Ressource
    ISSN: 2159-8274 , 2159-8290
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2020
    ZDB Id: 2607892-2
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 26, No. 13 ( 2020-07-01), p. 3287-3295
    Kurzfassung: Although first-line crizotinib treatment leads to clinical benefit in ROS1+ lung cancer, high prevalence of crizotinib-resistant ROS1-G2032R (ROS1G2032R) mutation and progression in the central nervous system (CNS) represents a therapeutic challenge. Here, we investigated the antitumor activity of repotrectinib, a novel next-generation ROS1/TRK/ALK-tyrosine kinase inhibitor (TKI) in ROS1+ patient-derived preclinical models. Experimental Design: Antitumor activity of repotrectinib was evaluated in ROS1+ patient-derived preclinical models including treatment-naïve and ROS1G2032R models and was further demonstrated in patients enrolled in an on-going phase I/II clinical trial (NCT03093116). Intracranial antitumor activity of repotrectinib was evaluated in a brain-metastasis mouse model. Results: Repotrectinib potently inhibited in vitro and in vivo tumor growth and ROS1 downstream signal in treatment-naïve YU1078 compared with clinically available crizotinib, ceritinib, and entrectinib. Despite comparable tumor regression between repotrectinib and lorlatinib in YU1078-derived xenograft model, repotrectinib markedly delayed the onset of tumor recurrence following drug withdrawal. Moreover, repotrectinib induced profound antitumor activity in the CNS with efficient blood–brain barrier penetrating properties. Notably, repotrectinib showed selective and potent in vitro and in vivo activity against ROS1G2032R. These findings were supported by systemic and intracranial activity of repotrectinib observed in patients enrolled in the on-going clinical trial. Conclusions: Repotrectinib is a novel next-generation ROS1-TKI with improved potency and selectivity against treatment-naïve and ROS1G2032R with efficient CNS penetration. Our findings suggest that repotrectinib can be effective both as first-line and after progression to prior ROS1-TKI.
    Materialart: Online-Ressource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2020
    ZDB Id: 1225457-5
    ZDB Id: 2036787-9
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 3071-3071
    Kurzfassung: The ROS1-rearranged non-small-cell lung cancer (NSCLC) is currently treated with ALK/ROS1 tyrosine kinase inhibitor (TKI) crizotinib as a first-line agent. However, resistance invariably develops and subsequent therapeutic option for overcoming them is limited. Therefore, we investigated the antitumor activity of several ROS1-TKIs in ROS1-positive patient-derived preclinical models. In this study, we established 6 patient-derived cells (PDCs) from patients with different types of ROS1 fusion partners as follows: treatment-naïve (YU1078, YU1082 and YU1083), crizotinib-resistant G2032R (YU1079) and crizotinib- or entrectinib- primary resistant (YU1081 and YU1085). Sanger sequencing and whole-exome sequencing were then conducted to determine the characteristics. The in vitro and in vivo antitumor activities of ROS1-TKIs in these PDCs were evaluated by comparing the survival, expression assay, duration of tumor re-emergence and ability to penetrate the blood-brain barrier (BBB). Repotrectinib potently inhibited cell proliferation and ROS1-downstream signaling pathways in YU1078 (CD74-ROS1). In YU1078-derived xenograft models, repotrectinib induced the marked tumor regression and delayed the duration of tumor re-emergence following drug withdrawal compared with crizotinib and entrectinib. Interestingly, both YU1081 (TPM3-ROS1) and YU1082 (SLC34A2-ROS1) with cross primary resistance to other clinically available ROS1-TKIs exhibited high sensitivity to repotrectinib in the concentration range of 100~200 nM. Immunoblot analysis demonstrated that repotrectinib completely suppressed the phosphorylation of ROS1 and STAT3. Notably, repotrectinib showed the selective and highly potency in vitro and in vivo against solvent-front mutant G2032R conferring crizotinib resistance. In G2032R-mutant xenograft models, the response of repotrectinib was still maintained until the end of the experiment (120 days) after drug withdrawal, but loratinib- or cabozantinib-treated tumors rapidly re-emerged. Moreover, repotrectinib induced profound tumor regression in brain metastasis model with excellent brain/plasma and tumor/brain area under the concentration-time curve value. As clinical proof of concept, the potent systemic and intracranial activity of ropotrectinib has been observed in patients who had progressed on prior TKIs or TKI-naïve patients, who were enrolled on-going phase 1/2 clinical trial (NCT03093116). Our finding indicated a superior antitumor activity of repotrectinib in both wild-type and mutant ROS1 fusion, providing a rationale that repotrectinib may be an effective subsequent or upfront treatment option for patients with ROS1-positive NSCLC who have relapsed on the available TKIs as well as TKI naïve, including patients with progressive CNS metastases. Citation Format: Dong Hwi Kim, Seok-Young Kim, Hyeong Seok Joo, You Won Lee, Han Na Kang, Min Hee Hong, Hye Ryun Kim, Byoung Chul Cho, Miran Yun. Repotrectinib demonstrates promising activities in ROS1 wild-type and solvent-front mutant lung cancer patients-derived preclinical models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3071.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2019
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 8 ( 2019-04-15), p. 2575-2587
    Kurzfassung: Given that osimertinib is the only approved third-generation EGFR-TKI against EGFR activating and resistant T790M mutated non–small cell lung cancer (NSCLC), additional mutant-selective inhibitors with a higher efficacy, especially for brain metastases, with favorable toxicity profile are still needed. In this study, we investigated the antitumor efficacy of YH25448, an oral, mutant-selective, irreversible third-generation EGFR-TKI in preclinical models. Experimental Design: Antitumor activity of YH25448 was investigated in vitro using mutant EGFR-expressing Ba/F3 cells and various lung cancer cell lines. In vivo antitumor efficacy, ability to penetrate the blood–brain barrier (BBB), and skin toxicity of YH25448 were examined and compared with those of osimertinib using cell lines and PDX model. Results: Compared with osimertinib, YH25448 showed a higher selectivity and potency in kinase assay and mutant EGFR-expressing Ba/F3 cells. In various cell line models harboring EGFR activating and T790M mutation, YH25448 effectively inhibited EGFR downstream signaling pathways, leading to cellular apoptosis. When compared in vivo at equimolar concentrations, YH25448 produced significantly better tumor regression than osimertinib. Importantly, YH25448 induced profound tumor regression in brain metastasis model with excellent brain/plasma and tumor/brain area under the concentration–time curve value. YH25448 rarely suppressed the levels of p-EGFR in hair follicles, leading to less keratosis than osimertinib in animal model. The potent systemic and intracranial activity of YH25448 has been shown in an ongoing phase I/II clinical trial for advanced EGFR T790M mutated NSCLC (NCT03046992). Conclusions: Our findings suggest that YH25448 is a promising third-generation EGFR inhibitor, which may be more effective and better tolerated than the currently approved osimertinib.
    Materialart: Online-Ressource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2019
    ZDB Id: 1225457-5
    ZDB Id: 2036787-9
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4790-4790
    Kurzfassung: EGFR mutated lung cancer shows approximately 10-15% of non-small cell lung cancer (NSCLC). Although the best therapeutic EGFR tyrosine kinase inhibitors (TKIs) targeting mutant EGFR, such as gefitinib and erlotinib, are used in the first line treatment of patients with advanced EGFR mutated NSCLC, the acquired resistance to the drugs usually appears in 10-12 months of therapy by the occurrence of a second EGFR mutation T790M. YH25448, a highly mutant-selective and irreversible 3rd generation EGFR TKI potently penetrating blood-brain barrier (BBB) penetration, targets both activating EGFR mutations Del19, L858R and T790M mutation while sparing wild type. In NSCLC cell lines and primary cancer cells from patients harboring EGFR mutations, YH25448 showed more potent inhibition of cancer cell growth and significantly increased tumor cell apoptosis compared to osimertinibs, which is one of 3rd generation EGFR TKIs. In vivo mouse model implanted with H1975 cells, YH25448 treatment at the once-daily showed a dramatic dose-dependent tumor regression in both subcutaneous and intracranial lesions with no abnormal signs such as skin keratosis shown in osimertinib-treated mice. Plasma half life of YH25448 was 5.9-6.8 hr and tumor to plasma AUC0-last ratio was 3.0-5.1 in tumor bearing mice. YH25448 also showed excellent penetration of the BBB, achieving CSF concentrations exceeding the IC50 value for pEGFR inhibition in the tumor-bearing mice. Taken together, these findings suggest important role for the further development of YH25448 as a novel therapeutic for the treatment of EGFR mutant-positive NSCLC patients with brain metastases. Citation Format: Jiyeon Yun, Min Hee Hong, Seok-Young Kim, Chae Won Park, So-Young Kim, Mi Ran Yun, Han Na Kang, Kyoung-Ho Pyo, Jong Sung Koh, Ho-Juhn Song, Young- Sung Lee, Se-Woong Oh, Soongyu Choi, Byoung-Chul Cho. YH25448, an irreversible 3rd generation EGFR TKI, exhibits superior anticancer effects with potent brain BBB penetration in NSCLC [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4790.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2018
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 5199-5199
    Kurzfassung: PURPOSE: Although EGFR exon20 insertion (ex20ins) mutations account for 4~12 % of EGFR mutant NSCLC patients, there is no effective and selectable anticancer drugs targeting ex20ins mutations so far due to various variant mutations in ex20ins mutations. Moreover, most EGFR ex20ins mutants show primary resistance to EGFR TKIs. JNJ-61186372 (JNJ-372), a bispecific antibody that targets the EGFR and cMet receptors, is currently being explored in a first-in-human study in patients with NSCLC. To better understand the mechanism of JNJ-372 activity in this patient population, we conducted preclinical studies exploring the activity of JNJ-372 in different EGFR ex20ins models. METHODS: To elucidate whether JNJ-372 has antitumor effect in EGFR ex20ins mutants via EGFR and c-MET inhibition, cell viability, western blot, cell cycle, colony formation assay, FACS analysis were performed in JNJ-372 treated BaF3 cells, PDCs, PDOs, and PDX expressing EGFR ex20ins mutation. For mouse tumor models, JNJ-372 was administered i.p. twice a week at 10 mg/kg or 30 mg/kg. Antibody dependent cellular cytotoxicity (ADCC) assay was assessed to figure out whether JNJ-372 had ADCC effects. Referenced patients with ex20ins disease were administered 1050 mg JNJ-372 i.v. weekly for the first 4-week cycle, then biweekly for each subsequent cycle RESULTS: JNJ-372 inhibited the growth of BaF3 cells, PDCs, and a PDO harboring a range of ex20ins, which were resistant to osimertinib and gefitinib. Mechanistic assays revealed the reduction of EGFR and cMet receptor levels and decreases in phospho-EGFR and c-Met, as well as inhibition of their downstream signaling pathways. Cleaved caspase-3 and BIMEL were upregulated at anti-proliferative doses, suggesting caspase-mediated cell death. JNJ-372 demonstrated corresponding antitumor activity in PDC and PDX models harboring different ex20ins; inhibition of signaling and engagement of the apoptotic pathway was confirmed in tumors of JNJ-372-treated mice. In PDCs with EGFR ex20ins mutation, we verified that JNJ-372 had a significant ADCC effect compared to EGFR antibody drug cetuximab. In the first-in-human trial, CT scans from two patients treated with JNJ-372 revealed reductions in tumor burden. A 58-year patient harboring H773delinsNPY showed -63% tumor reduction with progression-free survival of & gt; 20 months and a 48-year patient harboring S768_D770dup showed -38.9% tumor reduction. CONCLUSION: JNJ-372 drives antitumor activity in preclinical models of EGFR ex20ins, which have no therapeutic options in clinic, by decreasing EGFR and cMet receptor levels, inhibiting downstream signaling cascades, activating apoptotic signaling as well as ADCC. These results provide a promising therapeutic option to patients with EGFR ex20ins mutations and an understanding of the activity of JNJ-372 being observed in the first-in-human study. Citation Format: Jiyeon Yun, Han Na Kang, Soo-Hwan Lee, Seo-Yoon Jeong, Chae Won Park, Jae-Hwan Kim, Kyoung-Ho Pyo, Ji Min Lee, Seok-Young Kim, Min Hee Hong, Hye Ryun Kim, Meena Thayu, Joshua Curtin, Roland Knoblauch, Matthew Lorenzi, Byoung Chul Cho. JNJ-61186372, an EGFR-cMet bispecific antibody, in EGFR Exon 20 insertion-driven advanced non-small cell lung cancer (NSCLC) [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5199.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2020
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 64, No. 24 ( 2004-12-15), p. 8960-8967
    Kurzfassung: Although mechanisms of arsenic trioxide (As2O3)-induced cell death have been studied extensively in hematologic cancers, those in solid cancers have yet to be clearly defined. In this study, we showed that the translocation of apoptosis-inducing factor (AIF) from mitochondria to the nucleus is required for As2O3-induced cell death in human cervical cancer cells. We also showed that reactive oxygen species (ROS)-mediated poly(ADP-ribose) polymerase-1 (PARP-1) activation is necessary for AIF release from mitochondria. The treatment of human cervical cancer cells with As2O3 induces dissipation of mitochondrial membrane potential (Δψm), translocation of AIF from mitochondria to the nucleus, and subsequent cell death. Small interfering RNA targeting of AIF effectively protects cervical cancer cells against As2O3-induced cell death. As2O3 also induces an increase of intracellular ROS level and a marked activation of PARP-1. N-acetyl-l-cystein, a thiol-containing antioxidant, completely blocks As2O3-induced PARP-1 activation, Δψm loss, nuclear translocation of AIF from mitochondria, and the consequent cell death. Furthermore, pretreatment of 1,5-dihydroxyisoquinoline or 3,4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone, PARP-1 inhibitors, effectively attenuates the loss of Δψm, AIF release, and cell death. These data support a notion that ROS-mediated PARP-1 activation signals AIF release from mitochondria, resulting in activation of a caspase-independent pathway of cell death in solid tumor cells by As2O3 treatment.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2004
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 9
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 27, No. 15 ( 2021-08-01), p. 4397-4409
    Kurzfassung: Patient-derived organoids (PDO) of lung cancer has been recently introduced, reflecting the genomic landscape of lung cancer. However, clinical relevance of advanced lung adenocarcinoma organoids remains unknown. Here, we examined the ability of PDOs to predict clinical responses to targeted therapies in individual patients and to identify effective anticancer therapies for novel molecular targets. Experimental Design: Eighty-four organoids were established from patients with advanced lung adenocarcinoma. Formalin-fixed, paraffin-embedded tumor specimens from corresponding patients were analyzed by whole-exome sequencing (n = 12). Organoids were analyzed by whole-exome sequencing (n = 61) and RNA sequencing (n = 55). Responses to mono or combination targeted therapies were examined in organoids and organoid-derived xenografts. Results: PDOs largely retained somatic alterations including driver mutations of matching patient tumors. PDOs were able to recapitulate progression-free survival and objective responses of patients with non–small cell lung cancer receiving clinically approved tyrosine kinase inhibitors. PDOs recapitulated activity of therapeutic strategies under clinical investigation. YUO-071 harboring an EGFR exon 19 deletion and a BRAF G464A mutation and the matching patient responded to dabrafenib/trametinib combination therapy. YUO-004 and YUO-050 harboring an EGFR L747P mutation was sensitive to afatinib, consistent with the response in the matching patient of YUO-050. Furthermore, we utilized organoids to identify effective therapies for novel molecular targets by demonstrating the efficacy of poziotinib against ERBB2 exon 20 insertions and pralsetinib against RET fusions. Conclusions: We demonstrated translational relevance of PDOs in advanced lung adenocarcinoma. PDOs are an important diagnostic tool, which can assist clinical decision making and accelerate development of therapeutic strategies.
    Materialart: Online-Ressource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2021
    ZDB Id: 1225457-5
    ZDB Id: 2036787-9
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 1106-1106
    Kurzfassung: Background: EGFR-TKI is an established first-line therapy for NSCLC with activating EGFR mutations. Osimertinib, third-generation EGFR TKI, was investigated as a first-in-class drug, but lazertinib(YH25448) was also reported as an outstanding drug which had similar efficacy as osimertinib. Nevertheless, acquired resistance is inevitably developed by third-generation EGFR-TKIs in clinic. Even though EGFR C797S mutation has been identified as a major resistant mechanism, the remaining resistance mechanisms to these TKIs are largely unknown. Methods: To explore the mechanism of resistance to lazertinib, we firstly established drug-resistant cells to lazertinib in vitro using four NSCLC cells including the patient-derived cell line (PDC), patient-derived tumor xenograft cell line (PDTC), and ATCC cell lines. To analyze the changes in global genetic alterations and gene expression of established lazertinib-resistant cell lines(YH1R), whole exome sequencing (WES) and RNA sequencing (RNA-seq) were performed. The effect of EGFR/BRAF fusion was investigated in vitro using established YH1R cell line and various patient-derived tumor xenograft samples. Western blot study was used to investigate EGFR and MAPK cascade signaling pathway. We evaluated antitumor activity of MEK inhibitor and lazertinib combination treatment in lazertinib-resistant NSCLC cells with EGFR/BRAF fusion in vitro and in vivo. Results: RNA-seq data revealed a novel EGFR/BRAF fusion transcript (EGFR Exon 19-BRAF Exon 9, in-frame fusion) in lazertinib-resistant PC9GR_YH1R. The fusion gene consisted of EGFR (Exon 1-19) portion and BRAF kinase domain (Exon 9-18). The EGFR/BRAF fusion mRNA and protein were specifically upregulated in PC9GR_YH1R compared to parental cells. Intriguingly, we detected the EGFR/BRAF fusion gene expression in patient-derived xenografts (YHIM-1045 and YHIM-1035) obtained from patients who experienced acquired resistance to lazertinib. Compared with treatment with either single agent, combination treatment of lazertinib and MEK inhibitor, trametinib or selumetinib, was significantly effective at inhibiting cell proliferation as well as EGFR signaling pathways in PC9GR_YH1R in vitro and in vivo. Conclusion: We found a novel EGFR/BRAF fusion gene, as a key driver of acquired resistant mechanism of lazertinib, in NSCLC cell lines and patient-derived xenografts with acquired resistance to lazertinib. Combination treatment of lazertinib and MEK inhibitor showed a strong antitumor effect in lazertinib-acquired resistant NSCLCs, indicating that the combination therapy of EGFR and MEK inhibitors might be a promising therapeutic option for overcoming lazertinib-acquired resistant NSCLC patients in clinic. Citation Format: Seo-Yoon Jeong, Jiyeon Yun, San-Duk Yang, Soo-Hwan Lee, Sangbin Lim, Seok-Young Kim, Min Hee Hong, Sun Min Lim, Hye Ryun Kim, Hye Ryun Kim, Byoung Chul Cho. BRAF and EGFR fusion as a novel mechanism of resistance mechanism to Lazertinib, 3rd- generation EGFR-TKI, in EGFR-mutant NSCLC [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1106.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2021
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...