Pub Date : 2022-11-10eCollection Date: 2022-01-01DOI: 10.2147/LCTT.S383662
Danielle Brazel, Zhaohui Arter, Misako Nagasaka
KRASG12C is one of the most common oncogenes in non-small cell lung cancer (NSCLC) and is associated with a poor prognosis. Historically, KRAS mutations have been difficult to target due to lack of binding sites and exceptionally high affinity for guanosine triphosphate/guanosine diphosphate (GTP/GDP). Recently, KRASG12C selective inhibitors have shown promising results in Phase I/II studies. Here we discuss the mechanism of action, pharmacokinetic and pharmacodynamic properties, efficacy, and tolerability of adagrasib (MRTX849).
{"title":"A Long Overdue Targeted Treatment for KRAS Mutations in NSCLC: Spotlight on Adagrasib.","authors":"Danielle Brazel, Zhaohui Arter, Misako Nagasaka","doi":"10.2147/LCTT.S383662","DOIUrl":"https://doi.org/10.2147/LCTT.S383662","url":null,"abstract":"<p><p>KRAS<sup>G12C</sup> is one of the most common oncogenes in non-small cell lung cancer (NSCLC) and is associated with a poor prognosis. Historically, KRAS mutations have been difficult to target due to lack of binding sites and exceptionally high affinity for guanosine triphosphate/guanosine diphosphate (GTP/GDP). Recently, KRAS<sup>G12C</sup> selective inhibitors have shown promising results in Phase I/II studies. Here we discuss the mechanism of action, pharmacokinetic and pharmacodynamic properties, efficacy, and tolerability of adagrasib (MRTX849).</p>","PeriodicalId":18066,"journal":{"name":"Lung Cancer: Targets and Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2022-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/7e/c9/lctt-13-75.PMC9662012.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40708267","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-10-25eCollection Date: 2022-01-01DOI: 10.2147/LCTT.S385437
Shannon S Zhang, Sai-Hong Ignatius Ou
Osimertinib, a third-generation (3G) epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) is now considered the standard of care for the first-line (1L) treatment of advanced EGFR+ NSCLC due to statistically significant improved progression-free survival (PFS) and overall survival (OS) compared with first-generation (1G) treatment from the FLAURA trial. Recently two other 3G EGFR TKIs (aumolertinib and furmonertinib) have been approved in China for treatment of EGFR T790M+ NSCLC. Randomized Phase 3 trials of these two 3G EGFR TKIs have also demonstrated PFS over gefitinib respectively. Among these two Chinese home-grown, 3G EGFR TKIs, furmonertinib seems to most closely resemble osimertinib in terms of dosing regimen, efficacy and adverse events profile. In this article, we reviewed the clinical activity and adverse events of furmonertinib at 80 mg daily (approved dose), potential usage of 160 mg daily for CNS metastasis in EGFR+ NSCLC, and usage of 160 mg or 240 mg daily in EGFR exon20 insertion positive (EGFRex20ins+) NSCLC patients.
{"title":"Spotlight on Furmonertinib (Alflutinib, AST2818). The Swiss Army Knife (del19, L858R, T790M, Exon 20 Insertions, \"uncommon-G719X, S768I, L861Q\") Among the Third-Generation EGFR TKIs?","authors":"Shannon S Zhang, Sai-Hong Ignatius Ou","doi":"10.2147/LCTT.S385437","DOIUrl":"https://doi.org/10.2147/LCTT.S385437","url":null,"abstract":"<p><p>Osimertinib, a third-generation (3G) epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) is now considered the standard of care for the first-line (1L) treatment of advanced <i>EGFR</i>+ NSCLC due to statistically significant improved progression-free survival (PFS) and overall survival (OS) compared with first-generation (1G) treatment from the FLAURA trial. Recently two other 3G EGFR TKIs (aumolertinib and furmonertinib) have been approved in China for treatment of <i>EGFR</i> T790M+ NSCLC. Randomized Phase 3 trials of these two 3G EGFR TKIs have also demonstrated PFS over gefitinib respectively. Among these two Chinese home-grown, 3G EGFR TKIs, furmonertinib seems to most closely resemble osimertinib in terms of dosing regimen, efficacy and adverse events profile. In this article, we reviewed the clinical activity and adverse events of furmonertinib at 80 mg daily (approved dose), potential usage of 160 mg daily for CNS metastasis in <i>EGFR</i>+ NSCLC, and usage of 160 mg or 240 mg daily in <i>EGFR</i> exon20 insertion positive (<i>EGFRex20ins</i>+) NSCLC patients.</p>","PeriodicalId":18066,"journal":{"name":"Lung Cancer: Targets and Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2022-10-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/e0/7d/lctt-13-67.PMC9617553.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40659985","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-09-05eCollection Date: 2022-01-01DOI: 10.2147/LCTT.S335117
Joséphine Carpentier, Iuliia Pavlyk, Uma Mukherjee, Peter E Hall, Peter W Szlosarek
Arginine deprivation has gained increasing traction as a novel and safe antimetabolite strategy for the treatment of several hard-to-treat cancers characterised by a critical dependency on arginine. Small cell lung cancer (SCLC) displays marked arginine auxotrophy due to inactivation of the rate-limiting enzyme argininosuccinate synthetase 1 (ASS1), and as a consequence may be targeted with pegylated arginine deiminase or ADI-PEG20 (pegargiminase) and human recombinant pegylated arginases (rhArgPEG, BCT-100 and pegzilarginase). Although preclinical studies reveal that ASS1-deficient SCLC cell lines are highly sensitive to arginine-degrading enzymes, there is a clear disconnect with the clinic with minimal activity seen to date that may be due in part to patient selection. Recent studies have explored resistance mechanisms to arginine depletion focusing on tumor adaptation, such as ASS1 re-expression and autophagy, stromal cell inputs including macrophage infiltration, and tumor heterogeneity. Here, we explore how arginine deprivation may be combined strategically with novel agents to improve SCLC management by modulating resistance and increasing the efficacy of existing agents. Moreover, recent work has identified an intriguing role for targeting arginine in combination with PD-1/PD-L1 immune checkpoint inhibitors and clinical trials are in progress. Thus, future studies of arginine-depleting agents with chemoimmunotherapy, the current standard of care for SCLC, may lead to enhanced disease control and much needed improvements in long-term survival for patients.
{"title":"Arginine Deprivation in SCLC: Mechanisms and Perspectives for Therapy.","authors":"Joséphine Carpentier, Iuliia Pavlyk, Uma Mukherjee, Peter E Hall, Peter W Szlosarek","doi":"10.2147/LCTT.S335117","DOIUrl":"https://doi.org/10.2147/LCTT.S335117","url":null,"abstract":"<p><p>Arginine deprivation has gained increasing traction as a novel and safe antimetabolite strategy for the treatment of several hard-to-treat cancers characterised by a critical dependency on arginine. Small cell lung cancer (SCLC) displays marked arginine auxotrophy due to inactivation of the rate-limiting enzyme argininosuccinate synthetase 1 (ASS1), and as a consequence may be targeted with pegylated arginine deiminase or ADI-PEG20 (pegargiminase) and human recombinant pegylated arginases (rhArgPEG, BCT-100 and pegzilarginase). Although preclinical studies reveal that ASS1-deficient SCLC cell lines are highly sensitive to arginine-degrading enzymes, there is a clear disconnect with the clinic with minimal activity seen to date that may be due in part to patient selection. Recent studies have explored resistance mechanisms to arginine depletion focusing on tumor adaptation, such as ASS1 re-expression and autophagy, stromal cell inputs including macrophage infiltration, and tumor heterogeneity. Here, we explore how arginine deprivation may be combined strategically with novel agents to improve SCLC management by modulating resistance and increasing the efficacy of existing agents. Moreover, recent work has identified an intriguing role for targeting arginine in combination with PD-1/PD-L1 immune checkpoint inhibitors and clinical trials are in progress. Thus, future studies of arginine-depleting agents with chemoimmunotherapy, the current standard of care for SCLC, may lead to enhanced disease control and much needed improvements in long-term survival for patients.</p>","PeriodicalId":18066,"journal":{"name":"Lung Cancer: Targets and Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2022-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/a3/4b/lctt-13-53.PMC9462517.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"33466130","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-05-13eCollection Date: 2022-01-01DOI: 10.2147/LCTT.S360574
Danielle Brazel, Shannon Zhang, Misako Nagasaka
Mesenchymal-epithelial transition (MET) receptor tyrosine kinase is overexpressed, amplified, or mutated in 1-20% of NSCLC. MET dysregulation is associated with a poor prognosis. Recently, development of targeted therapies against MET exon 14 mutations has demonstrated efficacy and tolerability in early trials. Here we focus on tepotinib and capmatinib in regards to molecular characteristics, early preclinical and clinical data, and the emerging role in future studies and clinical practice.
间质-上皮转化(MET)受体酪氨酸激酶在 1-20% 的 NSCLC 中过度表达、扩增或突变。MET 失调与预后不良有关。最近,针对 MET 14 号外显子突变的靶向疗法在早期试验中显示出了疗效和耐受性。在此,我们将重点介绍特泊替尼(tepotinib)和卡马替尼(capmatinib)的分子特征、早期临床前和临床数据以及在未来研究和临床实践中的新作用。
{"title":"Spotlight on Tepotinib and Capmatinib for Non-Small Cell Lung Cancer with MET Exon 14 Skipping Mutation.","authors":"Danielle Brazel, Shannon Zhang, Misako Nagasaka","doi":"10.2147/LCTT.S360574","DOIUrl":"10.2147/LCTT.S360574","url":null,"abstract":"<p><p>Mesenchymal-epithelial transition (MET) receptor tyrosine kinase is overexpressed, amplified, or mutated in 1-20% of NSCLC. MET dysregulation is associated with a poor prognosis. Recently, development of targeted therapies against MET exon 14 mutations has demonstrated efficacy and tolerability in early trials. Here we focus on tepotinib and capmatinib in regards to molecular characteristics, early preclinical and clinical data, and the emerging role in future studies and clinical practice.</p>","PeriodicalId":18066,"journal":{"name":"Lung Cancer: Targets and Therapy","volume":null,"pages":null},"PeriodicalIF":5.1,"publicationDate":"2022-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9113513/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"76246502","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Abstract Tyrosine kinase inhibitors (TKIs) have become the preferred first line therapy for those patients with non-small cell lung cancer (NSCLC) harboring epidermal growth factor receptor (EGFR) mutations. Given superior progression free survival (PFS) and overall survival (OS) when compared to earlier generations, third generation EGFR TKIs have become the first choice therapy in many parts of the world. Even though multiple strategies are in development to target both “on-target” and “off-target” resistance, the continuation of EGFR TKIs at the time of progression remains a controversial topic. This commentary focuses on both the ”classic” clinical trials of IMpower150 and IMPRESS and compares them to the recently reported ORIENT-31 and ongoing MARIPOSA-2 to discuss the future therapeutic strategies in the setting of progression post-third generation EGFR TKIs.
{"title":"ORIENT-31 as the Sakigake “Charging Samurai” Born of IMpower150 but Will MARIPOSA-2 IMPRESS in the “Meiji Modernization” of Post-3G EGFR TKI Progression?","authors":"M. Nagasaka, S. Ou","doi":"10.2147/LCTT.S355503","DOIUrl":"https://doi.org/10.2147/LCTT.S355503","url":null,"abstract":"Abstract Tyrosine kinase inhibitors (TKIs) have become the preferred first line therapy for those patients with non-small cell lung cancer (NSCLC) harboring epidermal growth factor receptor (EGFR) mutations. Given superior progression free survival (PFS) and overall survival (OS) when compared to earlier generations, third generation EGFR TKIs have become the first choice therapy in many parts of the world. Even though multiple strategies are in development to target both “on-target” and “off-target” resistance, the continuation of EGFR TKIs at the time of progression remains a controversial topic. This commentary focuses on both the ”classic” clinical trials of IMpower150 and IMPRESS and compares them to the recently reported ORIENT-31 and ongoing MARIPOSA-2 to discuss the future therapeutic strategies in the setting of progression post-third generation EGFR TKIs.","PeriodicalId":18066,"journal":{"name":"Lung Cancer: Targets and Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2022-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"75702529","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Abstract The treatment paradigm for malignant pleural mesothelioma (MPM) has changed little in the last 18 years. Radical intent treatment, consisting of surgical resection, radiotherapy and chemotherapy, has been offered to a highly select few; however, there is little randomised evidence to validate this approach. Prior to 2020 chemotherapy with platinum and an anti-folate was the only intervention with randomised evidence to demonstrate improved overall survival (OS) in MPM. No systemic therapy had been demonstrated to improve OS in the second line setting until 2020. The publication of the Checkmate 743 trial in 2021 demonstrated a survival benefit of combination immunotherapy over standard chemotherapy in newly diagnosed patients with MPM. This finding was shortly followed by the CONFIRM trial which demonstrates a modest but significant survival benefit of second line nivolumab versus placebo in patients having previously received standard chemotherapy. The results of these trials, recent biomarker directed therapy and chemotherapy adjuncts are discussed within this review. The integration of immunotherapy for the few patients in whom radical surgical therapy is intended is currently the subject of clinical trials and offers the prospect of improving outcomes in this rare but devastating disease.
{"title":"An Update on Emerging Therapeutic Options for Malignant Pleural Mesothelioma","authors":"A. Davis, H. Ke, S. Kao, N. Pavlakis","doi":"10.2147/LCTT.S288535","DOIUrl":"https://doi.org/10.2147/LCTT.S288535","url":null,"abstract":"Abstract The treatment paradigm for malignant pleural mesothelioma (MPM) has changed little in the last 18 years. Radical intent treatment, consisting of surgical resection, radiotherapy and chemotherapy, has been offered to a highly select few; however, there is little randomised evidence to validate this approach. Prior to 2020 chemotherapy with platinum and an anti-folate was the only intervention with randomised evidence to demonstrate improved overall survival (OS) in MPM. No systemic therapy had been demonstrated to improve OS in the second line setting until 2020. The publication of the Checkmate 743 trial in 2021 demonstrated a survival benefit of combination immunotherapy over standard chemotherapy in newly diagnosed patients with MPM. This finding was shortly followed by the CONFIRM trial which demonstrates a modest but significant survival benefit of second line nivolumab versus placebo in patients having previously received standard chemotherapy. The results of these trials, recent biomarker directed therapy and chemotherapy adjuncts are discussed within this review. The integration of immunotherapy for the few patients in whom radical surgical therapy is intended is currently the subject of clinical trials and offers the prospect of improving outcomes in this rare but devastating disease.","PeriodicalId":18066,"journal":{"name":"Lung Cancer: Targets and Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2022-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83955144","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Recently, the ADAURA study demonstrated statistically significant improved disease-free survival (DFS) with adjuvant osimertinib in patients with resected stage IB-IIIA non-small cell lung cancer (NSCLC) harboring an epidermal growth factor receptor (EGFR) mutation. A consistent improvement in disease-free survival (DFS) was shown, regardless of whether or not patients received adjuvant chemotherapy. Given benefit seen with and without adjuvant chemotherapy, some clinicians may be tempted to forgo chemotherapy and only offer osimertinib post surgical resection. Would this approach be appropriate? Here we carefully dissect data from the ADAURA trial and review how this may fit into the existing evidence on the treatment of early stage NSCLC by discussing five themes, the study design of ADAURA, attempts on adjuvant tyrosine kinase inhibitors, prior studies to support adjuvant chemotherapy, how adjuvant chemotherapy was administered in ADAURA and consideration of toxicities.
{"title":"Deconstructing ADAURA. It is Not Yet Time to Forgo Platinum-based Adjuvant Chemotherapy in Resected Early Stage (IB-IIIA) EGFR-mutant NSCLC.","authors":"Danielle Brazel, Misako Nagasaka","doi":"10.2147/LCTT.S346922","DOIUrl":"https://doi.org/10.2147/LCTT.S346922","url":null,"abstract":"<p><p>Recently, the ADAURA study demonstrated statistically significant improved disease-free survival (DFS) with adjuvant osimertinib in patients with resected stage IB-IIIA non-small cell lung cancer (NSCLC) harboring an epidermal growth factor receptor (EGFR) mutation. A consistent improvement in disease-free survival (DFS) was shown, regardless of whether or not patients received adjuvant chemotherapy. Given benefit seen with and without adjuvant chemotherapy, some clinicians may be tempted to forgo chemotherapy and only offer osimertinib post surgical resection. Would this approach be appropriate? Here we carefully dissect data from the ADAURA trial and review how this may fit into the existing evidence on the treatment of early stage NSCLC by discussing five themes, the study design of ADAURA, attempts on adjuvant tyrosine kinase inhibitors, prior studies to support adjuvant chemotherapy, how adjuvant chemotherapy was administered in ADAURA and consideration of toxicities.</p>","PeriodicalId":18066,"journal":{"name":"Lung Cancer: Targets and Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/b4/b2/lctt-13-47.PMC9126226.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10254917","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Adjuvant cisplatin-based chemotherapy is considered the standard of care for resected stage IB (tumor ≥ 4m)-IIIA non-small cell lung cancer (NSCLC). The ADAURA trial is a randomized placebo-controlled Phase III trial that demonstrated statistically significant improved disease-free survival (DFS) with the use of 3-years of adjuvant osimertinib in resected stage IB-IIIA NSCLC harboring epidermal growth factor receptor (EGFR) del 19 or L858R mutations. Subgroup analysis revealed that the DFS improvement with adjuvant osimertinib is independent of adjuvant chemotherapy in the primary analysis. A recent follow-up report suggested that adjuvant cisplatin-based chemotherapy provided no additional 2-year DFS improvement on top of adjuvant osimertinib regardless of stage (IB, II, or IIIA) and minimal numerical DFS benefit in stage II or IIIA resected EGFR+ NSCLC for those patients who did not receive adjuvant osimertinib. Here, we argue that if clinicians adopt the use of 3 years of adjuvant osimertinib in resected early-stage EGFR+ NSCLC, there is no role for adjuvant platinum-based chemotherapy. The use of adjuvant chemotherapy was balanced between the osimertinib and the placebo arms by stage even though adjuvant chemotherapy was not one of the three stratification factors (del 19 vs L858R; Stage IA vs II vs III; Asians versus non-Asian) in ADAURA. There may be a potential role of adjuvant cisplatin/vinorelbine in a small subgroup of EGFR+ NSCLC patients whose tumor harbors retinoblastoma (RB) gene alterations but requires further investigation.
以顺铂为基础的辅助化疗被认为是IB期(肿瘤≥4m)-IIIA期非小细胞肺癌(NSCLC)切除的标准治疗方案。ADAURA试验是一项随机安慰剂对照III期试验,该试验显示,在切除的具有表皮生长因子受体(EGFR) del 19或L858R突变的IB-IIIA期NSCLC中,使用3年辅助奥西替尼可显著提高无病生存率(DFS)。亚组分析显示,在初步分析中,辅助奥希替尼对DFS的改善与辅助化疗无关。最近的一项随访报告表明,在辅助奥西替尼的基础上,辅助顺铂化疗没有提供额外的2年DFS改善,无论分期(IB, II或IIIA),对于那些没有接受辅助奥西替尼的II期或IIIA期EGFR+ NSCLC切除患者,DFS的数值最小。在这里,我们认为,如果临床医生在切除的早期EGFR+ NSCLC中采用3年的辅助奥希替尼,那么基于铂的辅助化疗就没有作用。辅助化疗的使用在奥希替尼组和安慰剂组之间的分期是平衡的,即使辅助化疗不是三个分层因素之一(del 19 vs L858R;IA期vs II期vs III期;亚洲人与非亚洲人)的ADAURA。在一小部分EGFR+ NSCLC患者(其肿瘤含有视网膜母细胞瘤(RB)基因改变)中,顺铂/维诺瑞滨可能具有潜在的辅助作用,但需要进一步研究。
{"title":"Deconstructing ADAURA: It is Time to Forgo Adjuvant Platinum-Based Chemotherapy in Resected IB-IIIA <i>EGFR+</i> NSCLC (Except with <i>RB</i> Alterations?) When Adopting Adjuvant Osimertinib.","authors":"Shannon S Zhang, Sai-Hong Ignatius Ou","doi":"10.2147/LCTT.S358902","DOIUrl":"https://doi.org/10.2147/LCTT.S358902","url":null,"abstract":"<p><p>Adjuvant cisplatin-based chemotherapy is considered the standard of care for resected stage IB (tumor ≥ 4m)-IIIA non-small cell lung cancer (NSCLC). The ADAURA trial is a randomized placebo-controlled Phase III trial that demonstrated statistically significant improved disease-free survival (DFS) with the use of 3-years of adjuvant osimertinib in resected stage IB-IIIA NSCLC harboring epidermal growth factor receptor (<i>EGFR</i>) del 19 or L858R mutations. Subgroup analysis revealed that the DFS improvement with adjuvant osimertinib is independent of adjuvant chemotherapy in the primary analysis. A recent follow-up report suggested that adjuvant cisplatin-based chemotherapy provided no additional 2-year DFS improvement on top of adjuvant osimertinib regardless of stage (IB, II, or IIIA) and minimal numerical DFS benefit in stage II or IIIA resected <i>EGFR+</i> NSCLC for those patients who did not receive adjuvant osimertinib. Here, we argue that if clinicians adopt the use of 3 years of adjuvant osimertinib in resected early-stage <i>EGFR+</i> NSCLC, there is no role for adjuvant platinum-based chemotherapy. The use of adjuvant chemotherapy was balanced between the osimertinib and the placebo arms by stage even though adjuvant chemotherapy was not one of the three stratification factors (del 19 vs L858R; Stage IA vs II vs III; Asians versus non-Asian) in ADAURA. There may be a potential role of adjuvant cisplatin/vinorelbine in a small subgroup of <i>EGFR+</i> NSCLC patients whose tumor harbors retinoblastoma (<i>RB</i>) gene alterations but requires further investigation.</p>","PeriodicalId":18066,"journal":{"name":"Lung Cancer: Targets and Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/6a/a6/lctt-13-23.PMC9057228.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10615149","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-12-02eCollection Date: 2021-01-01DOI: 10.2147/LCTT.S272518
Natalie I Vokes, Jianjun Zhang
Non-small cell lung cancer (NSCLC) that presents with multiple lung tumors (MLTs) poses a challenge to accurate staging and prognosis. MLTs that arise as clonally related secondary metastases from a common primary are higher stage and often require adjuvant chemotherapy or may in fact be incurable stage IV lesions. Conversely, MLTs that represent distinct primaries have a better prognosis and may be overtreated if inappropriately classified as related secondaries. Historically, pathologic and radiographic criteria were used to distinguish between primary and secondary MLTs; however, the advent of genomic profiling has demonstrated limitations to these historic classification systems. In this review, we discuss the use of molecular profiling to distinguish between primary and secondary lung cancers, with a focus on the insights gleaned from whole exome sequencing (WES) analyses. While WES is not yet feasible in routine clinical practice, WES studies have helped elucidate the clonal relationship between primary and secondary lung cancers and provide important context for the application of targeted sequencing panel-based analyses.
非小细胞肺癌(NSCLC)伴有多发性肺肿瘤(MLT),给准确分期和预后带来了挑战。从一个共同的原发灶继发转移而来的克隆相关的多发性肺肿瘤分期较高,通常需要辅助化疗,或实际上可能是无法治愈的 IV 期病变。相反,代表不同原发灶的 MLT 预后较好,如果被不恰当地归类为相关的继发灶,则可能会过度治疗。历史上,病理学和放射学标准被用于区分原发性和继发性 MLT;然而,基因组分析的出现表明了这些历史性分类系统的局限性。在这篇综述中,我们将讨论如何利用分子图谱来区分原发性和继发性肺癌,重点是全外显子组测序(WES)分析所获得的启示。虽然全外显子组测序在常规临床实践中还不可行,但全外显子组测序研究有助于阐明原发性和继发性肺癌之间的克隆关系,并为基于靶向测序面板分析的应用提供了重要的背景。
{"title":"The Role of Whole Exome Sequencing in Distinguishing Primary and Secondary Lung Cancers.","authors":"Natalie I Vokes, Jianjun Zhang","doi":"10.2147/LCTT.S272518","DOIUrl":"10.2147/LCTT.S272518","url":null,"abstract":"<p><p>Non-small cell lung cancer (NSCLC) that presents with multiple lung tumors (MLTs) poses a challenge to accurate staging and prognosis. MLTs that arise as clonally related secondary metastases from a common primary are higher stage and often require adjuvant chemotherapy or may in fact be incurable stage IV lesions. Conversely, MLTs that represent distinct primaries have a better prognosis and may be overtreated if inappropriately classified as related secondaries. Historically, pathologic and radiographic criteria were used to distinguish between primary and secondary MLTs; however, the advent of genomic profiling has demonstrated limitations to these historic classification systems. In this review, we discuss the use of molecular profiling to distinguish between primary and secondary lung cancers, with a focus on the insights gleaned from whole exome sequencing (WES) analyses. While WES is not yet feasible in routine clinical practice, WES studies have helped elucidate the clonal relationship between primary and secondary lung cancers and provide important context for the application of targeted sequencing panel-based analyses.</p>","PeriodicalId":18066,"journal":{"name":"Lung Cancer: Targets and Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2021-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/ca/0d/lctt-12-139.PMC8648100.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39793525","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-12-02eCollection Date: 2021-01-01DOI: 10.2147/LCTT.S337861
Danielle Brazel, Misako Nagasaka
Non-small cell lung cancer (NSCLC) patients demonstrating sensitizing oncogenic driver mutations have derived clinical benefit from targeted therapy. EGFR mutations constitutively activate the signaling pathway, leading to prosurvival and antiapoptotic signals. Classic sensitizing EGFR mutations, such as exon 19 deletions and exon 21 L858R point mutations, respond well to tyrosine kinase inhibitors (TKIs). On the other hand, EGFR exon 20 in-frame insertions are observed in 4-12% of EGFR-mutated NSCLC and are resistant to targeted therapy with TKIs. In May 2021, the Federal Drug Administration (FDA) provided accelerated approval to amivantamab (Rybrevant) in adults with locally advanced or metastatic NSCLC with EGFR exon 20 insertion mutations after treatment with platinum-based chemotherapy. Here, we discuss properties of amivantamab, clinical trial results, and management of patients with EGFR exon 20 insertion mutated NSCLC.
{"title":"Spotlight on Amivantamab (JNJ-61186372) for EGFR Exon 20 Insertions Positive Non-Small Cell Lung Cancer.","authors":"Danielle Brazel, Misako Nagasaka","doi":"10.2147/LCTT.S337861","DOIUrl":"10.2147/LCTT.S337861","url":null,"abstract":"<p><p>Non-small cell lung cancer (NSCLC) patients demonstrating sensitizing oncogenic driver mutations have derived clinical benefit from targeted therapy. EGFR mutations constitutively activate the signaling pathway, leading to prosurvival and antiapoptotic signals. Classic sensitizing EGFR mutations, such as exon 19 deletions and exon 21 L858R point mutations, respond well to tyrosine kinase inhibitors (TKIs). On the other hand, EGFR exon 20 in-frame insertions are observed in 4-12% of EGFR-mutated NSCLC and are resistant to targeted therapy with TKIs. In May 2021, the Federal Drug Administration (FDA) provided accelerated approval to amivantamab (Rybrevant) in adults with locally advanced or metastatic NSCLC with EGFR exon 20 insertion mutations after treatment with platinum-based chemotherapy. Here, we discuss properties of amivantamab, clinical trial results, and management of patients with EGFR exon 20 insertion mutated NSCLC.</p>","PeriodicalId":18066,"journal":{"name":"Lung Cancer: Targets and Therapy","volume":null,"pages":null},"PeriodicalIF":5.1,"publicationDate":"2021-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/21/88/lctt-12-133.PMC8648093.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39793524","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}