首页 > 最新文献

Cancer最新文献

英文 中文
Shedding LIGHT on the changing landscape of poly-ADP ribose polymerase inhibitors in ovarian cancer
IF 6.1 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-07 DOI: 10.1002/cncr.35800
Melissa K. Frey MD, MS, Joyce F. Liu MD, MPH
{"title":"Shedding LIGHT on the changing landscape of poly-ADP ribose polymerase inhibitors in ovarian cancer","authors":"Melissa K. Frey MD, MS, Joyce F. Liu MD, MPH","doi":"10.1002/cncr.35800","DOIUrl":"https://doi.org/10.1002/cncr.35800","url":null,"abstract":"","PeriodicalId":138,"journal":{"name":"Cancer","volume":"131 8","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143787080","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Worse health quality outcomes found with endocrine therapy after breast-conserving surgery
IF 6.1 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-03 DOI: 10.1002/cncr.35791
Mary Beth Nierengarten

Endocrine therapy following breast-conserving surgery in older women with low-risk, early-stage breast cancer was associated with a greater reduction in health-related quality of life (HRQOL) than radiation therapy according to the interim analysis of the EUROPA trial presented at the San Antonio Breast Cancer Symposium1 and published in The Lancet Oncology.2

The EUROPA trial is a noninferiority, phase 3 study in which 731 women (with a final accrual target of 926 patients) aged 70 years or older with stage I, luminal A–like breast cancer were randomized to a single-modality treatment with either radiotherapy (n = 365) or endocrine therapy (n = 366). The study compared the effects on HRQOL and ipsilateral breast tumor recurrence. Changes in HRQOL from the baseline to 2 years were assessed with the global health status scale of the 30-item European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire.

Results of the preplanned interim analysis, which included 207 patients (104 received radiotherapy, and 103 received endocrine therapy), showed a significant reduction in HRQOL in the patients treated with endocrine therapy versus radiotherapy.

At 2 years, patients treated with endocrine therapy had a mean change from the baseline in global health status of –9.79 (95% CI, –14.45 to –5.13; p < .0001), whereas the mean change for those treated with radiotherapy was –3.40 (95% CI, –7.82 to 1.03; p = .13). The adjusted mean difference between the treatments was 6.39 (95% CI, 0.14–12.65; p = .04), with radiotherapy favored.

Patients treated with radiotherapy also had less frequent treatment-related adverse events than those treated with endocrine therapy (67% vs. 85%). The most common grade 3–4 adverse events were experienced by patients in the endocrinology group (none in the radiotherapy group) and included arthralgia (7%), pelvic organ prolapse (3%), fatigue/hot flashes/myalgia (2%), and bone pain/fractures (2%). Serious adverse events were similar between the two treatment groups.

The lead author of the study, Icro Meattini, MD, an associate professor in the Department of Experimental and Clinical Biomedical Sciences “M. Serio” at the University of Florence in Italy, says that the study shows that single-modality therapy has promise as a noninferior alternative to endocrine therapy for improved HRQOL.

“These findings highlight the potential for personalized treatment approaches that align with patient preferences, comorbidities, and treatment tolerability,” he says, adding that the findings “could influence shared decision-making and reduce overtreatment in a subset of older patients with favorable tumor biology.”

Although the interim results are encouraging, he says that the trial will continue as planned to ensure a robust data set for the final analysis.

{"title":"Worse health quality outcomes found with endocrine therapy after breast-conserving surgery","authors":"Mary Beth Nierengarten","doi":"10.1002/cncr.35791","DOIUrl":"https://doi.org/10.1002/cncr.35791","url":null,"abstract":"<p>Endocrine therapy following breast-conserving surgery in older women with low-risk, early-stage breast cancer was associated with a greater reduction in health-related quality of life (HRQOL) than radiation therapy according to the interim analysis of the EUROPA trial presented at the San Antonio Breast Cancer Symposium<span><sup>1</sup></span> and published in <i>The Lancet Oncology</i>.<span><sup>2</sup></span></p><p>The EUROPA trial is a noninferiority, phase 3 study in which 731 women (with a final accrual target of 926 patients) aged 70 years or older with stage I, luminal A–like breast cancer were randomized to a single-modality treatment with either radiotherapy (<i>n</i> = 365) or endocrine therapy (<i>n</i> = 366). The study compared the effects on HRQOL and ipsilateral breast tumor recurrence. Changes in HRQOL from the baseline to 2 years were assessed with the global health status scale of the 30-item European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire.</p><p>Results of the preplanned interim analysis, which included 207 patients (104 received radiotherapy, and 103 received endocrine therapy), showed a significant reduction in HRQOL in the patients treated with endocrine therapy versus radiotherapy.</p><p>At 2 years, patients treated with endocrine therapy had a mean change from the baseline in global health status of –9.79 (95% CI, –14.45 to –5.13; <i>p</i> &lt; .0001), whereas the mean change for those treated with radiotherapy was –3.40 (95% CI, –7.82 to 1.03; <i>p</i> = .13). The adjusted mean difference between the treatments was 6.39 (95% CI, 0.14–12.65; <i>p</i> = .04), with radiotherapy favored.</p><p>Patients treated with radiotherapy also had less frequent treatment-related adverse events than those treated with endocrine therapy (67% vs. 85%). The most common grade 3–4 adverse events were experienced by patients in the endocrinology group (none in the radiotherapy group) and included arthralgia (7%), pelvic organ prolapse (3%), fatigue/hot flashes/myalgia (2%), and bone pain/fractures (2%). Serious adverse events were similar between the two treatment groups.</p><p>The lead author of the study, Icro Meattini, MD, an associate professor in the Department of Experimental and Clinical Biomedical Sciences “M. Serio” at the University of Florence in Italy, says that the study shows that single-modality therapy has promise as a noninferior alternative to endocrine therapy for improved HRQOL.</p><p>“These findings highlight the potential for personalized treatment approaches that align with patient preferences, comorbidities, and treatment tolerability,” he says, adding that the findings “could influence shared decision-making and reduce overtreatment in a subset of older patients with favorable tumor biology.”</p><p>Although the interim results are encouraging, he says that the trial will continue as planned to ensure a robust data set for the final analysis.</p>","PeriodicalId":138,"journal":{"name":"Cancer","volume":"131 7","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cncr.35791","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143761855","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Key findings highlighted in the first annual cancer report from the American College of Surgeons
IF 6.1 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-03 DOI: 10.1002/cncr.35789
Mary Beth Nierengarten
<p>The American College of Surgeons Cancer Program developed its first annual cancer report from the National Cancer Database (NCDB) 2021 Participant User File highlighting data on adult cancer diagnoses, patient demographics, and treatments.<span><sup>1</sup></span> Data on cancer cases from the NCDB are collected from approximately 1500 Commission on Cancer facilities and account for nearly 74% of newly diagnosed cancer cases nationally.</p><p>The report features key demographic information for the five most common cancers: breast, prostate, lung, colon, and melanoma. It also includes an in-depth look at treatments and outcomes of three select cancers: breast, pancreatic, and colorectal. Future annual reports will focus on other cancer types.</p><p>The senior author of the study, Judy C. Boughey, MD, a surgical oncologist and division chair of breast and melanoma surgical oncology at the Mayo Clinic in Rochester, Minnesota, says that the report was conducted to offer cancer providers information on cancer cases from the NCDB and to highlight the data over time along with patient outcomes.</p><p>“Overall, this is a report from a large database demonstrating the management of patients with certain solid tumors for physicians to be able to refer to regarding factors influencing outcome, current practice patterns, as well as hospital admission rates and morbidities associated with cancer surgery,” she says.</p><p>“This data is available for providers for their knowledge and to share with their patients,” she adds.</p><p>Overall, 1,328,131 adult cancers were diagnosed in 2021; this was a slight increase from the 1,277,700 adult cancers diagnosed in 2020. The data showed more cancer diagnoses in females than males (54.3% vs. 45.7%); in part, this shows the large number of women diagnosed with breast cancer (composing one-fifth or 19.9% of all adult cancers in 2021).</p><p>The data also include overall survival for patients diagnosed through 2020. Overall, 5-year survival was 63.99% for all tumor sites, with markedly better 5-year survival rates in women versus men (68.01% vs. 59.36%).</p><p>In 2021, 264,095 adults (99.2% women and 0.8% men) were diagnosed with breast cancer, as reported by 1276 NCDB-participating facilities; this figure represents an estimated 81.9% of all breast cancers diagnosed in the United States.<span><sup>2</sup></span> The median age at diagnosis was 63 years; most patients were White (74%), followed by Black (12.4%), Hispanic (6.1%), and Asian (5%). The stages at diagnosis were similar to what had been seen since 2018, with most patients presenting with stage I (47%) or stage II disease (24%); these stages were followed by ductal carcinoma in situ (18%), stage III disease (5%), and stage IV disease (5%). Most patients underwent lumpectomy (66.4%) followed by radiation therapy (80.6%). Among patients with stage I–III breast cancers, 37.4% received chemotherapy, and 14.4% received immunotherapy. Among those with ER+ tumors, 86
{"title":"Key findings highlighted in the first annual cancer report from the American College of Surgeons","authors":"Mary Beth Nierengarten","doi":"10.1002/cncr.35789","DOIUrl":"https://doi.org/10.1002/cncr.35789","url":null,"abstract":"&lt;p&gt;The American College of Surgeons Cancer Program developed its first annual cancer report from the National Cancer Database (NCDB) 2021 Participant User File highlighting data on adult cancer diagnoses, patient demographics, and treatments.&lt;span&gt;&lt;sup&gt;1&lt;/sup&gt;&lt;/span&gt; Data on cancer cases from the NCDB are collected from approximately 1500 Commission on Cancer facilities and account for nearly 74% of newly diagnosed cancer cases nationally.&lt;/p&gt;&lt;p&gt;The report features key demographic information for the five most common cancers: breast, prostate, lung, colon, and melanoma. It also includes an in-depth look at treatments and outcomes of three select cancers: breast, pancreatic, and colorectal. Future annual reports will focus on other cancer types.&lt;/p&gt;&lt;p&gt;The senior author of the study, Judy C. Boughey, MD, a surgical oncologist and division chair of breast and melanoma surgical oncology at the Mayo Clinic in Rochester, Minnesota, says that the report was conducted to offer cancer providers information on cancer cases from the NCDB and to highlight the data over time along with patient outcomes.&lt;/p&gt;&lt;p&gt;“Overall, this is a report from a large database demonstrating the management of patients with certain solid tumors for physicians to be able to refer to regarding factors influencing outcome, current practice patterns, as well as hospital admission rates and morbidities associated with cancer surgery,” she says.&lt;/p&gt;&lt;p&gt;“This data is available for providers for their knowledge and to share with their patients,” she adds.&lt;/p&gt;&lt;p&gt;Overall, 1,328,131 adult cancers were diagnosed in 2021; this was a slight increase from the 1,277,700 adult cancers diagnosed in 2020. The data showed more cancer diagnoses in females than males (54.3% vs. 45.7%); in part, this shows the large number of women diagnosed with breast cancer (composing one-fifth or 19.9% of all adult cancers in 2021).&lt;/p&gt;&lt;p&gt;The data also include overall survival for patients diagnosed through 2020. Overall, 5-year survival was 63.99% for all tumor sites, with markedly better 5-year survival rates in women versus men (68.01% vs. 59.36%).&lt;/p&gt;&lt;p&gt;In 2021, 264,095 adults (99.2% women and 0.8% men) were diagnosed with breast cancer, as reported by 1276 NCDB-participating facilities; this figure represents an estimated 81.9% of all breast cancers diagnosed in the United States.&lt;span&gt;&lt;sup&gt;2&lt;/sup&gt;&lt;/span&gt; The median age at diagnosis was 63 years; most patients were White (74%), followed by Black (12.4%), Hispanic (6.1%), and Asian (5%). The stages at diagnosis were similar to what had been seen since 2018, with most patients presenting with stage I (47%) or stage II disease (24%); these stages were followed by ductal carcinoma in situ (18%), stage III disease (5%), and stage IV disease (5%). Most patients underwent lumpectomy (66.4%) followed by radiation therapy (80.6%). Among patients with stage I–III breast cancers, 37.4% received chemotherapy, and 14.4% received immunotherapy. Among those with ER+ tumors, 86","PeriodicalId":138,"journal":{"name":"Cancer","volume":"131 7","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cncr.35789","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143761856","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Longer progression-free survival with targeted therapy for patients with metastatic breast cancer
IF 6.1 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-03 DOI: 10.1002/cncr.35790
Mary Beth Nierengarten
<p>Select patients with metastatic breast cancer who received trastuzumab deruxtecan (T-DXd) after endocrine therapy had significantly longer progression-free survival (PFS) than patients treated with chemotherapy according to the results of the DESTINY-Breast06 trial presented at the San Antonio Breast Cancer Symposium and published in <i>The New England Journal of Medicine</i>.<span><sup>1, 2</sup></span></p><p>Specifically, patients with hormone receptor–positive (HR+) metastatic breast cancer with low human epidermal growth factor receptor 2 (HER2) who were treated with T-DXd had a median PFS of 13.2 months versus 8.1 months for those treated with chemotherapy. This represents a 38% reduction in the risk of disease progression or death for those treated with T-DXd (hazard ratio, 0.62; 95% CI, 0.52–0.75; <i>p</i> < .001).</p><p>Similarly improved PFS was seen for patients with ultralow HER2 expression (a score of 0 on an immunohistochemical [IHC] analysis with membrane staining), although the main outcome of the study was PFS among patients with HER2-low disease (a score of 1+ or 2+ on an IHC analysis and negative results on in situ hybridization).</p><p>DESTINY-Breast06 is a phase 3, multicenter, open-label trial in which 866 patients with HR+ metastatic cancer with low or ultralow HER2 expression who had received one or more lines of endocrine-based therapy (and no previous chemotherapy) were randomized to T-DXd (<i>n</i> = 436) or chemotherapy (<i>n</i> = 430) according to the physician’s choice. T-DXd, an antibody–drug conjugate, was administered every 3 weeks. Among the patients, 713 had HER2-low disease, and 153 had HER2-ultralow disease.</p><p>The results provide more clarity on the sequencing of T-DXd in patients with metastatic breast cancer with low HER2 expression. Although T-DXd has shown efficacy in this setting when delivered after chemotherapy, its efficacy when delivered earlier after endocrine-based therapy is unknown.</p><p>Giuseppe Curigliano, MD, PhD, senior deputy director at the European Institute of Oncology in Milan, says that the study is important because it shows that T-DXd could become the new standard of care for patients with HR+, HER2-low, or HER2-ultralow metastatic breast cancer. “These data underscore the potential for treatment with T-DXd across the spectrum of HR+ breast cancer, further redefining the treatment of metastatic breast cancer,” he says.</p><p>Oladapo Yeku, MD, PhD, associate editor of <i>NEJM Evidence</i>, said in an audio interview that the “study supports the hypothesis that antibody–drug conjugates, such as trastuzumab deruxtecan, are effective in earlier lines of treatment even when patients have not been exposed to conventional cytotoxic chemotherapy.”<span><sup>3</sup></span></p><p>When he was asked what factors should be considered when one is considering using T-DXd or chemotherapy in this setting, he cited the different types of toxicities associated with each type of treatment.</p><
{"title":"Longer progression-free survival with targeted therapy for patients with metastatic breast cancer","authors":"Mary Beth Nierengarten","doi":"10.1002/cncr.35790","DOIUrl":"https://doi.org/10.1002/cncr.35790","url":null,"abstract":"&lt;p&gt;Select patients with metastatic breast cancer who received trastuzumab deruxtecan (T-DXd) after endocrine therapy had significantly longer progression-free survival (PFS) than patients treated with chemotherapy according to the results of the DESTINY-Breast06 trial presented at the San Antonio Breast Cancer Symposium and published in &lt;i&gt;The New England Journal of Medicine&lt;/i&gt;.&lt;span&gt;&lt;sup&gt;1, 2&lt;/sup&gt;&lt;/span&gt;&lt;/p&gt;&lt;p&gt;Specifically, patients with hormone receptor–positive (HR+) metastatic breast cancer with low human epidermal growth factor receptor 2 (HER2) who were treated with T-DXd had a median PFS of 13.2 months versus 8.1 months for those treated with chemotherapy. This represents a 38% reduction in the risk of disease progression or death for those treated with T-DXd (hazard ratio, 0.62; 95% CI, 0.52–0.75; &lt;i&gt;p&lt;/i&gt; &lt; .001).&lt;/p&gt;&lt;p&gt;Similarly improved PFS was seen for patients with ultralow HER2 expression (a score of 0 on an immunohistochemical [IHC] analysis with membrane staining), although the main outcome of the study was PFS among patients with HER2-low disease (a score of 1+ or 2+ on an IHC analysis and negative results on in situ hybridization).&lt;/p&gt;&lt;p&gt;DESTINY-Breast06 is a phase 3, multicenter, open-label trial in which 866 patients with HR+ metastatic cancer with low or ultralow HER2 expression who had received one or more lines of endocrine-based therapy (and no previous chemotherapy) were randomized to T-DXd (&lt;i&gt;n&lt;/i&gt; = 436) or chemotherapy (&lt;i&gt;n&lt;/i&gt; = 430) according to the physician’s choice. T-DXd, an antibody–drug conjugate, was administered every 3 weeks. Among the patients, 713 had HER2-low disease, and 153 had HER2-ultralow disease.&lt;/p&gt;&lt;p&gt;The results provide more clarity on the sequencing of T-DXd in patients with metastatic breast cancer with low HER2 expression. Although T-DXd has shown efficacy in this setting when delivered after chemotherapy, its efficacy when delivered earlier after endocrine-based therapy is unknown.&lt;/p&gt;&lt;p&gt;Giuseppe Curigliano, MD, PhD, senior deputy director at the European Institute of Oncology in Milan, says that the study is important because it shows that T-DXd could become the new standard of care for patients with HR+, HER2-low, or HER2-ultralow metastatic breast cancer. “These data underscore the potential for treatment with T-DXd across the spectrum of HR+ breast cancer, further redefining the treatment of metastatic breast cancer,” he says.&lt;/p&gt;&lt;p&gt;Oladapo Yeku, MD, PhD, associate editor of &lt;i&gt;NEJM Evidence&lt;/i&gt;, said in an audio interview that the “study supports the hypothesis that antibody–drug conjugates, such as trastuzumab deruxtecan, are effective in earlier lines of treatment even when patients have not been exposed to conventional cytotoxic chemotherapy.”&lt;span&gt;&lt;sup&gt;3&lt;/sup&gt;&lt;/span&gt;&lt;/p&gt;&lt;p&gt;When he was asked what factors should be considered when one is considering using T-DXd or chemotherapy in this setting, he cited the different types of toxicities associated with each type of treatment.&lt;/p&gt;&lt;","PeriodicalId":138,"journal":{"name":"Cancer","volume":"131 7","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cncr.35790","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143761854","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Advances in the treatment of KRASG12C mutant non–small cell lung cancer
IF 6.1 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1002/cncr.35783
Khvaramze Shaverdashvili MD, PhD, Timothy F. Burns MD, PhD

Kirsten rat sarcoma (KRAS) is one of the most frequently mutated oncogenic drivers in metastatic non–small cell lung cancer (NSCLC). The development of selective, covalent KRAS G12C (KRASG12C) inhibitors represents a breakthrough in the treatment for KRASG12C mutant NSCLC, but the durability of response and efficacy of these inhibitors are limited by the rapid emergence of drug resistance and their ability to only bind KRASG12C in the guanosine diphosphate-bound form. Importantly, co-occurring gene alterations, including KEAP1, STK11, and CDKN2A, may affect prognosis and response to therapies, including immunotherapy and KRASG12C inhibitors. New therapeutic approaches are needed to both delay and overcome treatment resistance. Moreover, developing KRAS inhibitors with novel mechanisms of action and alternative allele specificities is necessary to overcome emerging on-target resistance mechanisms to KRASG12C inhibitors. A literature search was performed using PubMed, the Food and Drug Administration website, and Google search. The inclusive dates in the literature search were between 1982 and July 2024. In this article, the authors reviewed the disease prevalence, biology and therapeutic options, including specific KRASG12C inhibitors and new pan-KRAS therapeutic agents for KRASG12C mutant NSCLC. KRAS inhibitor resistance mechanisms, treatment strategies, and multi-targeted treatment approaches are also discussed.

{"title":"Advances in the treatment of KRASG12C mutant non–small cell lung cancer","authors":"Khvaramze Shaverdashvili MD, PhD,&nbsp;Timothy F. Burns MD, PhD","doi":"10.1002/cncr.35783","DOIUrl":"https://doi.org/10.1002/cncr.35783","url":null,"abstract":"<p><i>Kirsten rat sarcoma</i> (<i>KRAS</i>) is one of the most frequently mutated oncogenic drivers in metastatic non–small cell lung cancer (NSCLC). The development of selective, covalent KRAS G12C (KRAS<sup>G12C</sup>) inhibitors represents a breakthrough in the treatment for <i>KRAS</i><sup><i>G12C</i></sup> mutant NSCLC, but the durability of response and efficacy of these inhibitors are limited by the rapid emergence of drug resistance and their ability to only bind KRAS<sup>G12C</sup> in the guanosine diphosphate-bound form. Importantly, co-occurring gene alterations, including <i>KEAP1</i>, <i>STK11</i>, and <i>CDKN2A</i>, may affect prognosis and response to therapies, including immunotherapy and KRAS<sup>G12C</sup> inhibitors. New therapeutic approaches are needed to both delay and overcome treatment resistance. Moreover, developing KRAS inhibitors with novel mechanisms of action and alternative allele specificities is necessary to overcome emerging on-target resistance mechanisms to KRAS<sup>G12C</sup> inhibitors. A literature search was performed using PubMed, the Food and Drug Administration website, and Google search. The inclusive dates in the literature search were between 1982 and July 2024. In this article, the authors reviewed the disease prevalence, biology and therapeutic options, including specific KRAS<sup>G12C</sup> inhibitors and new pan-KRAS therapeutic agents for <i>KRAS</i><sup><i>G12C</i></sup> mutant NSCLC. KRAS inhibitor resistance mechanisms, treatment strategies, and multi-targeted treatment approaches are also discussed.</p>","PeriodicalId":138,"journal":{"name":"Cancer","volume":"131 S1","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cncr.35783","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143749388","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Update advances in anaplastic lymphoma kinase-positive non–small cell lung cancer treatment
IF 6.1 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1002/cncr.35786
Beatriz E. Jimenez Munarriz MD, Sam Khan MBChB, PhD, Yuchen Li MD, Nadia Ghazali B.Med, Geoffrey Liu MSc, MD

Lung cancer is the leading cause of cancer death worldwide. However, since the discovery and development of targeted therapies and immune checkpoint inhibitors, it has been a dramatically improved in survival outcomes and quality of life. Anaplastic lymphoma kinase (ALK) rearrangements occur in approximately 5% of patients with advanced non–small cell lung cancer (NSCLC), and the introduction of ALK tyrosine kinase inhibitors (TKIs) has changed the treatment paradigm. Although head-to-head randomized controlled trials for late generation inhibitors are lacking, the authors will review the current available options, treatments efficacy, safety profiles, ALK mechanisms of resistance, as well as new promising treatments in this field.

{"title":"Update advances in anaplastic lymphoma kinase-positive non–small cell lung cancer treatment","authors":"Beatriz E. Jimenez Munarriz MD,&nbsp;Sam Khan MBChB, PhD,&nbsp;Yuchen Li MD,&nbsp;Nadia Ghazali B.Med,&nbsp;Geoffrey Liu MSc, MD","doi":"10.1002/cncr.35786","DOIUrl":"https://doi.org/10.1002/cncr.35786","url":null,"abstract":"<p>Lung cancer is the leading cause of cancer death worldwide. However, since the discovery and development of targeted therapies and immune checkpoint inhibitors, it has been a dramatically improved in survival outcomes and quality of life. Anaplastic lymphoma kinase (<i>ALK</i>) rearrangements occur in approximately 5% of patients with advanced non–small cell lung cancer (NSCLC), and the introduction of ALK tyrosine kinase inhibitors (TKIs) has changed the treatment paradigm. Although head-to-head randomized controlled trials for late generation inhibitors are lacking, the authors will review the current available options, treatments efficacy, safety profiles, ALK mechanisms of resistance, as well as new promising treatments in this field.</p>","PeriodicalId":138,"journal":{"name":"Cancer","volume":"131 S1","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143749387","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Multicancer early detection testing: Guidance for primary care discussions with patients
IF 6.1 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1002/cncr.35823
Richard M. Hoffman MD, MPH, Andrew M. D. Wolf MD, Sana Raoof MD, PhD, Carmen E. Guerra MD, MSCE, Timothy R. Church PhD, Elena B. Elkin PhD, MPA, Ruth D. Etzioni PhD, Ya-Chen Tina Shih PhD, Steven J. Skates PhD, Deana Manassaram-Baptiste PhD, MPH, Robert A. Smith PhD

Multicancer early detection (MCED) tests are an emerging technology for cancer screening. MCED tests can detect cancer signals from multiple cancers concurrently in biological samples such as blood, urine, saliva, or other bodily fluids. Some tests can suggest the most likely cancer origin, whereas others report cancer detected somewhere in the body. Although some MCED tests are currently commercially available, none are approved by the Food and Drug Administration or endorsed by any clinical practice guideline or recommendation. Most insurance companies do not currently cover MCED testing. MCED tests have not yet been evaluated for safety and effectiveness in randomized controlled trials. Because patients already are asking for MCED test prescriptions or for interpretation of results from tests acquired elsewhere, clinicians should be prepared to discuss what is known about the benefits, risks, and uncertainties of MCED testing, including performance characteristics in screening populations and preferred follow-up strategies for positive test results. At this time, clinicians should not feel obligated to initiate discussions about MCED testing with their patients. However, clinicians should engage patients who inquire about getting tested or previous MCED test results in shared decision-making, and take the opportunity to offer and help patients complete age- and sex-appropriate guideline-recommended cancer screenings. In this article, the current evidence and issues around MCED testing are summarized, and a framework for shared decision-making discussions is provided.

{"title":"Multicancer early detection testing: Guidance for primary care discussions with patients","authors":"Richard M. Hoffman MD, MPH,&nbsp;Andrew M. D. Wolf MD,&nbsp;Sana Raoof MD, PhD,&nbsp;Carmen E. Guerra MD, MSCE,&nbsp;Timothy R. Church PhD,&nbsp;Elena B. Elkin PhD, MPA,&nbsp;Ruth D. Etzioni PhD,&nbsp;Ya-Chen Tina Shih PhD,&nbsp;Steven J. Skates PhD,&nbsp;Deana Manassaram-Baptiste PhD, MPH,&nbsp;Robert A. Smith PhD","doi":"10.1002/cncr.35823","DOIUrl":"https://doi.org/10.1002/cncr.35823","url":null,"abstract":"<p>Multicancer early detection (MCED) tests are an emerging technology for cancer screening. MCED tests can detect cancer signals from multiple cancers concurrently in biological samples such as blood, urine, saliva, or other bodily fluids. Some tests can suggest the most likely cancer origin, whereas others report cancer detected somewhere in the body. Although some MCED tests are currently commercially available, none are approved by the Food and Drug Administration or endorsed by any clinical practice guideline or recommendation. Most insurance companies do not currently cover MCED testing. MCED tests have not yet been evaluated for safety and effectiveness in randomized controlled trials. Because patients already are asking for MCED test prescriptions or for interpretation of results from tests acquired elsewhere, clinicians should be prepared to discuss what is known about the benefits, risks, and uncertainties of MCED testing, including performance characteristics in screening populations and preferred follow-up strategies for positive test results. At this time, clinicians should not feel obligated to initiate discussions about MCED testing with their patients. However, clinicians should engage patients who inquire about getting tested or previous MCED test results in shared decision-making, and take the opportunity to offer and help patients complete age- and sex-appropriate guideline-recommended cancer screenings. In this article, the current evidence and issues around MCED testing are summarized, and a framework for shared decision-making discussions is provided.</p>","PeriodicalId":138,"journal":{"name":"Cancer","volume":"131 7","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cncr.35823","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143749550","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting HER2 in lung cancers: Evolving treatment landscape and drug development strategies
IF 6.1 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1002/cncr.35780
Daniel Reinhorn MD, Mor Moskovitz MD, William D. Tap MD, Bob T. Li MD, PHD, MPH

ERBB2 (HER2) alterations, including mutations, amplifications, and overexpression are emerging therapeutic targets in non–small cell lung cancer (NSCLC). Despite recent advancements, standard first-line therapy remains chemotherapy with or without immunotherapy. Several therapies targeting HER2 are under development and have been evaluated in clinical trials with inconsistent efficacy, including monoclonal antibodies, tyrosine kinase inhibitors, and antibody–drug conjugates. A major landmark was recently reached when trastuzumab deruxtecan (T-DXd), an antibody–drug conjugate, became the first Food and Drug Administration (FDA)-approved therapy for pretreated HER2-mutant NSCLC, following the promising efficacy demonstrated in the DESTINY-Lung trials. Furthermore, T-DXd has shown efficacy across various tumor types harboring HER2 alterations in the DESTINY-PanTumor trials, leading to its recent pan-tumor FDA approval for HER2-positive solid tumors, highlighting the potential of tumor-agnostic drug development strategies. In this review, the authors describe the different HER2 alterations and their clinical consequences, including their impact on prognosis and response to standard therapies. They provide an up-to-date overview of the current treatment landscape and add a comprehensive review of pivotal and ongoing clinical trials of HER2-targeted therapies, including tumor-agnostic drug development strategies.

{"title":"Targeting HER2 in lung cancers: Evolving treatment landscape and drug development strategies","authors":"Daniel Reinhorn MD,&nbsp;Mor Moskovitz MD,&nbsp;William D. Tap MD,&nbsp;Bob T. Li MD, PHD, MPH","doi":"10.1002/cncr.35780","DOIUrl":"https://doi.org/10.1002/cncr.35780","url":null,"abstract":"<p>ERBB2 (HER2) alterations, including mutations, amplifications, and overexpression are emerging therapeutic targets in non–small cell lung cancer (NSCLC). Despite recent advancements, standard first-line therapy remains chemotherapy with or without immunotherapy. Several therapies targeting HER2 are under development and have been evaluated in clinical trials with inconsistent efficacy, including monoclonal antibodies, tyrosine kinase inhibitors, and antibody–drug conjugates. A major landmark was recently reached when trastuzumab deruxtecan (T-DXd), an antibody–drug conjugate, became the first Food and Drug Administration (FDA)-approved therapy for pretreated HER2-mutant NSCLC, following the promising efficacy demonstrated in the DESTINY-Lung trials. Furthermore, T-DXd has shown efficacy across various tumor types harboring HER2 alterations in the DESTINY-PanTumor trials, leading to its recent pan-tumor FDA approval for HER2-positive solid tumors, highlighting the potential of tumor-agnostic drug development strategies. In this review, the authors describe the different HER2 alterations and their clinical consequences, including their impact on prognosis and response to standard therapies. They provide an up-to-date overview of the current treatment landscape and add a comprehensive review of pivotal and ongoing clinical trials of HER2-targeted therapies, including tumor-agnostic drug development strategies.</p>","PeriodicalId":138,"journal":{"name":"Cancer","volume":"131 S1","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143749390","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
BRAF in non–small cell lung cancer: From molecular mechanisms to clinical practice
IF 6.1 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1002/cncr.35781
Claudia Parisi MD, David Planchard MD, PhD

V-Raf murine sarcoma viral oncogene homolog B (BRAF) mutations are found in up to 4% of patients with non–small cell lung cancer (NSCLC). Approximately 2% of advanced NSCLC cases harbor a BRAF V600E (class I) mutation. Because targeted therapies inhibiting BRAF (e.g., dabrafenib and encorafenib) and MEK (trametinib and binimetinib) are associated with improved outcomes as first- or second-line treatment for BRAF V600E–mutant NSCLC, both European Society for Medical Oncology and National Comprehensive Cancer Network guidelines recommend testing for the BRAF V600E oncogenic driver at the time of diagnosis. In recent years, the treatment landscape of this molecular subgroup has seen great development. Different therapeutic strategies including anti–programmed death ligand 1 antibodies and kinase inhibitors have been assessed thus far, with novel agents (e.g., pan-BRAF inhibitors) and therapeutic associations underway in preclinical and clinical trials. This review describes the current understanding of the BRAF clinicopathologic role in NSCLC, with a special focus on published trials assessing currently approved therapies. Mechanisms of drug resistance and future perspectives on the therapeutic approach of BRAF-deregulated NSCLC are also summarized.

{"title":"BRAF in non–small cell lung cancer: From molecular mechanisms to clinical practice","authors":"Claudia Parisi MD,&nbsp;David Planchard MD, PhD","doi":"10.1002/cncr.35781","DOIUrl":"https://doi.org/10.1002/cncr.35781","url":null,"abstract":"<p>V-Raf murine sarcoma viral oncogene homolog B (<i>BRAF</i>) mutations are found in up to 4% of patients with non–small cell lung cancer (NSCLC). Approximately 2% of advanced NSCLC cases harbor a <i>BRAF</i> V600E (class I) mutation. Because targeted therapies inhibiting <i>BRAF</i> (e.g., dabrafenib and encorafenib) and <i>MEK</i> (trametinib and binimetinib) are associated with improved outcomes as first- or second-line treatment for <i>BRAF</i> V600E–mutant NSCLC, both European Society for Medical Oncology and National Comprehensive Cancer Network guidelines recommend testing for the <i>BRAF</i> V600E oncogenic driver at the time of diagnosis. In recent years, the treatment landscape of this molecular subgroup has seen great development. Different therapeutic strategies including anti–programmed death ligand 1 antibodies and kinase inhibitors have been assessed thus far, with novel agents (e.g., pan-BRAF inhibitors) and therapeutic associations underway in preclinical and clinical trials. This review describes the current understanding of the <i>BRAF</i> clinicopathologic role in NSCLC, with a special focus on published trials assessing currently approved therapies. Mechanisms of drug resistance and future perspectives on the therapeutic approach of <i>BRAF</i>-deregulated NSCLC are also summarized.</p>","PeriodicalId":138,"journal":{"name":"Cancer","volume":"131 S1","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143749459","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting ROS1 rearrangements in non–small cell lung cancer: Current insights and future directions
IF 6.1 2区 医学 Q1 ONCOLOGY Pub Date : 2025-04-02 DOI: 10.1002/cncr.35784
Antoine Desilets MD, MSc, Matteo Repetto MD, Soo-Ryum Yang MD, Alexander Drilon MD

ROS1 rearrangements define a molecular subset of non–small cell lung cancer (NSCLC) by accounting for 1%–2% of cases. Targeted therapy with ROS1 tyrosine kinase inhibitors (TKIs) has significantly improved the outcomes for these patients. First-generation inhibitors, such as crizotinib and entrectinib, have demonstrated impressive efficacy, with objective response rates exceeding 60%–70%. However, the emergence of resistance mechanisms, including solvent-front mutations such as ROS1 G2032R, and limited blood–brain barrier penetration have limited the long-term efficacy of early-generation agents. Next-generation TKIs, including lorlatinib, taletrectinib, and repotrectinib, have been developed to overcome these challenges. These agents show enhanced central nervous system (CNS) penetration and activity against on-target ROS1 resistance mutations. Repotrectinib, a potent, CNS-penetrant ROS1 inhibitor, has demonstrated superior activity in both TKI-naive and -resistant tumors, including those harboring the G2032R mutation. Zidesamtinib, a highly selective next-generation ROS1 inhibitor, further addresses TRK-mediated off-target neurological toxicities seen with prior agents, and is poised to offer improved tolerability. Ongoing research is focused on optimizing sequencing strategies for ROS1 inhibitors and exploring combination approaches to prevent or overcome resistance. In addition, the development of novel diagnostic tools, including RNA-based next-generation sequencing, has enhanced the detection of functional ROS1 fusions by ensuring that patients with actionable mutations receive appropriate targeted therapies. These advances highlight the evolving landscape of treatment for ROS1-positive NSCLC, with the aim of maximizing long-term survival and quality of life.

{"title":"Targeting ROS1 rearrangements in non–small cell lung cancer: Current insights and future directions","authors":"Antoine Desilets MD, MSc,&nbsp;Matteo Repetto MD,&nbsp;Soo-Ryum Yang MD,&nbsp;Alexander Drilon MD","doi":"10.1002/cncr.35784","DOIUrl":"https://doi.org/10.1002/cncr.35784","url":null,"abstract":"<p><i>ROS1</i> rearrangements define a molecular subset of non–small cell lung cancer (NSCLC) by accounting for 1%–2% of cases. Targeted therapy with ROS1 tyrosine kinase inhibitors (TKIs) has significantly improved the outcomes for these patients. First-generation inhibitors, such as crizotinib and entrectinib, have demonstrated impressive efficacy, with objective response rates exceeding 60%–70%. However, the emergence of resistance mechanisms, including solvent-front mutations such as <i>ROS1</i> G2032R, and limited blood–brain barrier penetration have limited the long-term efficacy of early-generation agents. Next-generation TKIs, including lorlatinib, taletrectinib, and repotrectinib, have been developed to overcome these challenges. These agents show enhanced central nervous system (CNS) penetration and activity against on-target <i>ROS1</i> resistance mutations. Repotrectinib, a potent, CNS-penetrant ROS1 inhibitor, has demonstrated superior activity in both TKI-naive and -resistant tumors, including those harboring the G2032R mutation. Zidesamtinib, a highly selective next-generation ROS1 inhibitor, further addresses TRK-mediated off-target neurological toxicities seen with prior agents, and is poised to offer improved tolerability. Ongoing research is focused on optimizing sequencing strategies for ROS1 inhibitors and exploring combination approaches to prevent or overcome resistance. In addition, the development of novel diagnostic tools, including RNA-based next-generation sequencing, has enhanced the detection of functional <i>ROS1</i> fusions by ensuring that patients with actionable mutations receive appropriate targeted therapies. These advances highlight the evolving landscape of treatment for <i>ROS1</i>-positive NSCLC, with the aim of maximizing long-term survival and quality of life.</p>","PeriodicalId":138,"journal":{"name":"Cancer","volume":"131 S1","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143749460","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1