首页 > 最新文献

CA: A Cancer Journal for Clinicians最新文献

英文 中文
Reviewer acknowledgement 2024 审稿人确认 2024
IF 254.7 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-18 DOI: 10.3322/caac.21866

In order to maintain the high standards of CA’s content, the Editors of CA rely on the knowledge and dedication of many experts in deciding which topics to pursue, which manuscripts to publish, and what modifications to make to ensure medical and scientific accuracy and suitability for our readers. We thank our Associate Editors and our Editorial Advisory Board, who continue to provide these services for us time and time again.

We are also greatly indebted to the effort and expertise of the following individuals for reviewing manuscripts for the journal from July 1, 2023, to June 30, 2024. These individuals go beyond expectations by consistently and expeditiously delivering comprehensive, discerning reviews. Peer review is an essential component of scholarly publishing, and we sincerely appreciate the time and expertise volunteered by these participants.

Dina Amin

Eric Bernicker

Chao Cao

Raymond Chan

Paul Daeninck

Fernando Diaz

Tanya Dorff

Georges Gebrael

Samir Hanash

Vida Henderson

Hormuzd Katki

Shumei Kato

Amir Khan

Allison J. Lazard

Mark Lewis

Kim Margolin

Joaquin Mateo

Justin Moyers

Maria Pisu

Gwendolyn Quinn

Paul Riviere

Jason Sicklick

Zbyslaw Sondka

Conor Steuer

Kristine Swartz

Randy Sweis

Peter Yu

Xue Qin Yu

为了保持CA内容的高标准,CA的编辑们依靠许多专家的知识和奉献精神来决定哪些主题需要探讨、哪些稿件需要发表、哪些稿件需要修改以确保医学和科学的准确性并适合我们的读者。我们感谢我们的副主编和编辑顾问委员会,他们一次又一次地为我们提供这些服务。我们还非常感谢以下人员在 2023 年 7 月 1 日至 2024 年 6 月 30 日期间为本刊审稿所付出的努力和专业知识。这些人始终如一地迅速提供全面、独到的审稿意见,超出了我们的预期。Dina AminEric BernickerChao CaoRaymond ChanPaul DaeninckFernando DiazTanya DorffGeorges GebraelSamir HanashVida HendersonHormuzd KatkiShumei KatoAmir KhanAllison J.LazardMark LewisKim MargolinJoaquin MateoJustin MoyersMaria PisuGwendolyn QuinnPaul RiviereJason SicklickZbyslaw SondkaConor SteuerKristine SwartzRandy SweisPeter Yu薛勤宇
{"title":"Reviewer acknowledgement 2024","authors":"","doi":"10.3322/caac.21866","DOIUrl":"https://doi.org/10.3322/caac.21866","url":null,"abstract":"<p>In order to maintain the high standards of <i>CA</i>’s content, the Editors of <i>CA</i> rely on the knowledge and dedication of many experts in deciding which topics to pursue, which manuscripts to publish, and what modifications to make to ensure medical and scientific accuracy and suitability for our readers. We thank our Associate Editors and our Editorial Advisory Board, who continue to provide these services for us time and time again.</p>\u0000<p>We are also greatly indebted to the effort and expertise of the following individuals for reviewing manuscripts for the journal from July 1, 2023, to June 30, 2024. These individuals go beyond expectations by consistently and expeditiously delivering comprehensive, discerning reviews. Peer review is an essential component of scholarly publishing, and we sincerely appreciate the time and expertise volunteered by these participants.</p>\u0000<p>Dina Amin</p>\u0000<p>Eric Bernicker</p>\u0000<p>Chao Cao</p>\u0000<p>Raymond Chan</p>\u0000<p>Paul Daeninck</p>\u0000<p>Fernando Diaz</p>\u0000<p>Tanya Dorff</p>\u0000<p>Georges Gebrael</p>\u0000<p>Samir Hanash</p>\u0000<p>Vida Henderson</p>\u0000<p>Hormuzd Katki</p>\u0000<p>Shumei Kato</p>\u0000<p>Amir Khan</p>\u0000<p>Allison J. Lazard</p>\u0000<p>Mark Lewis</p>\u0000<p>Kim Margolin</p>\u0000<p>Joaquin Mateo</p>\u0000<p>Justin Moyers</p>\u0000<p>Maria Pisu</p>\u0000<p>Gwendolyn Quinn</p>\u0000<p>Paul Riviere</p>\u0000<p>Jason Sicklick</p>\u0000<p>Zbyslaw Sondka</p>\u0000<p>Conor Steuer</p>\u0000<p>Kristine Swartz</p>\u0000<p>Randy Sweis</p>\u0000<p>Peter Yu</p>\u0000<p>Xue Qin Yu</p>","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":null,"pages":null},"PeriodicalIF":254.7,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142237013","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Cancer disparities for LGBTQ+ patients identified more fully 更全面地确定 LGBTQ+ 患者的癌症差异。
IF 503.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-02 DOI: 10.3322/caac.21861
Mike Fillon
<p>It has been widely reported that patients who identify as LGBTQ+ (lesbian, gay, bisexual, transgender, queer, or other gender-diverse characteristic) have more health risks than the cisgender and/or heterosexual population. According to previous studies, most of the disparity has been attributed to the minority stress theory: Members of these communities disproportionally experience discrimination, and this results in mistrust in medical settings—further increasing stress.</p><p>Regarding cancer specifically, these society-derived stressors have been reported to lead to lower rates of timely screening, higher rates of infection with cancer-causing viruses, and higher rates of health risk behaviors—increasing the potential risk for various cancers in the LGBTQ+ community. Another issue builds on the aforementioned minority stress theory, which can result in avoidance because of the fear that a health care provider will refuse to care for them. Importantly, the LGBTQ+ communities are diverse, and cancer incidences may differ within specific gender identities and/or sexual orientations (SOs). Because of insufficient details from previous studies, accurate data regarding cancer incidence in specific groups have been lacking.</p><p>A study appearing in <i>Cancer</i> (doi: 10.1002/cncr.35356) adds new evidence of the disproportional cancer burden faced by sexual minoritized people. Study author Aimee K. Huang, MD, MPH, a junior faculty member at Massachusetts General Hospital and Harvard Medical School in Boston, Massachusetts, says that most prior studies relied on indirect approximations of incidence and prevalence. “However, for studies that were able to directly measure incidence, the scopes of their investigations were often limited to the most common cancers, unidimensional SO measurements, or had other methodological challenges due to data limitations,” she says.</p><p>For the study, researchers culled SO and cancer diagnosis data (from 1989 to 2017) from the Nurses’ Health Study II (NHSII), a longitudinal cohort of 101,543 nurses across the United States. The mean ages and race/ethnicity compositions were similar across all the groups.</p><p>The primary outcome was the self-disclosed and electronic health record–verified incidences of cancer among four different sexual minority groups: heterosexual with a past same-sex attraction/behavior/identity (<i>n</i> = 5034), mostly heterosexual (<i>n</i> = 1825), bisexual (<i>n</i> = 394), and lesbian (<i>n</i> = 996). These groups were compared to a “reference” group that self-identified as lifelong heterosexual (<i>n</i> = 93,294). The researchers also determined the case numbers, incidence rates, and age-adjusted incidence rate ratios (aIRRs) of 21 site-specific cancers for each group. Using aIRRs, they compared incidence rates between the reference group and the four SO subgroups.</p><p>The researchers reported that the cancer incidence rate (cases per 100,000 person-years) was highest for those
据广泛报道,被认定为 LGBTQ+(女同性恋、男同性恋、双性恋、变性人、同性恋者或其他具有不同性别特征的人)的患者比同性和/或异性恋人群面临更多的健康风险。根据以往的研究,这种差异大多归因于少数群体压力理论:具体到癌症,据报道,这些来自社会的压力导致 LGBTQ+ 群体及时筛查的比例较低、感染致癌病毒的比例较高以及健康风险行为的比例较高,从而增加了他们罹患各种癌症的潜在风险。另一个问题建立在上述少数群体压力理论的基础上,由于担心医疗服务提供者会拒绝为他们提供医疗服务,这可能会导致他们回避。重要的是,LGBTQ+ 群体具有多样性,在特定的性别认同和/或性倾向(SOs)中,癌症发病率可能有所不同。癌症》(Cancer)杂志刊登的一项研究(doi: 10.1002/cncr.35356)为性取向少数群体面临的不成比例的癌症负担提供了新的证据。该研究的作者、马萨诸塞州波士顿马萨诸塞州总医院和哈佛大学医学院青年教师 Aimee K. Huang(医学博士、公共卫生硕士)说,之前的大多数研究都依赖于对发病率和流行率的间接近似值。她说:"然而,对于那些能够直接测量发病率的研究,其调查范围往往局限于最常见的癌症、单维SO测量,或者由于数据限制而面临其他方法上的挑战,"在这项研究中,研究人员从护士健康研究II(NHSII)中收集了SO和癌症诊断数据(从1989年到2017年),NHSII是一个由全美101,543名护士组成的纵向队列。主要结果是四个不同的性少数群体自我披露和电子健康记录核实的癌症发病率:过去有同性吸引/行为/身份的异性恋(n = 5034)、大部分为异性恋(n = 1825)、双性恋(n = 394)和女同性恋(n = 996)。这些群体与自我认同为终身异性恋的 "参照 "群体(n = 93 294)进行了比较。研究人员还确定了每个群体的病例数、发病率以及 21 种特定部位癌症的年龄调整发病率比(aIRRs)。研究人员报告说,自我描述为女同性恋者的癌症发病率(每 10 万人年的病例数)最高(516 例)。参照组参与者的发病率为 428 例,略低于有过去同性吸引/伴侣/身份的异性恋女性(449 例)和大多数异性恋人群(439 例)。那些自我描述为双性恋的人构成了最小的群体,研究人员认为无法得出准确的结论。他们发现,女同性恋者被诊断出三种癌症的几率大约是男同性恋者的两倍:甲状腺癌(aIRR,1.87)、基底细胞癌(aIRR,1.85)和非霍奇金淋巴瘤(aIRR,2.13)。研究发现,性少数群体妇女被诊断出肺癌的风险较低,尽管之前的研究报告称这些群体中吸烟和患肺癌的比例较高。研究还发现女同性恋者中没有肺癌病例,并发现有同性性行为史的异性恋女性肺癌发病率较低。研究作者认为,NHSII 队列中的这些发现可能是由于这一护理人群的吸烟率低于普通公众。"他们写道:"因此,我们发现性少数群体女性的肺癌发病率较低,但病例数较少,对此应谨慎解释。(纽约州罗切斯特市罗切斯特大学医学系多元化、公平与包容副主任、医学博士 Chunkit Fung 说,美国的这项大型纵向研究非常重要,因为它的详细报告显示,性少数群体女性的所有癌症发病率都高于异性恋女性。他说,一个特别重要的发现是,女同性恋者的癌症负担最重。
{"title":"Cancer disparities for LGBTQ+ patients identified more fully","authors":"Mike Fillon","doi":"10.3322/caac.21861","DOIUrl":"10.3322/caac.21861","url":null,"abstract":"&lt;p&gt;It has been widely reported that patients who identify as LGBTQ+ (lesbian, gay, bisexual, transgender, queer, or other gender-diverse characteristic) have more health risks than the cisgender and/or heterosexual population. According to previous studies, most of the disparity has been attributed to the minority stress theory: Members of these communities disproportionally experience discrimination, and this results in mistrust in medical settings—further increasing stress.&lt;/p&gt;&lt;p&gt;Regarding cancer specifically, these society-derived stressors have been reported to lead to lower rates of timely screening, higher rates of infection with cancer-causing viruses, and higher rates of health risk behaviors—increasing the potential risk for various cancers in the LGBTQ+ community. Another issue builds on the aforementioned minority stress theory, which can result in avoidance because of the fear that a health care provider will refuse to care for them. Importantly, the LGBTQ+ communities are diverse, and cancer incidences may differ within specific gender identities and/or sexual orientations (SOs). Because of insufficient details from previous studies, accurate data regarding cancer incidence in specific groups have been lacking.&lt;/p&gt;&lt;p&gt;A study appearing in &lt;i&gt;Cancer&lt;/i&gt; (doi: 10.1002/cncr.35356) adds new evidence of the disproportional cancer burden faced by sexual minoritized people. Study author Aimee K. Huang, MD, MPH, a junior faculty member at Massachusetts General Hospital and Harvard Medical School in Boston, Massachusetts, says that most prior studies relied on indirect approximations of incidence and prevalence. “However, for studies that were able to directly measure incidence, the scopes of their investigations were often limited to the most common cancers, unidimensional SO measurements, or had other methodological challenges due to data limitations,” she says.&lt;/p&gt;&lt;p&gt;For the study, researchers culled SO and cancer diagnosis data (from 1989 to 2017) from the Nurses’ Health Study II (NHSII), a longitudinal cohort of 101,543 nurses across the United States. The mean ages and race/ethnicity compositions were similar across all the groups.&lt;/p&gt;&lt;p&gt;The primary outcome was the self-disclosed and electronic health record–verified incidences of cancer among four different sexual minority groups: heterosexual with a past same-sex attraction/behavior/identity (&lt;i&gt;n&lt;/i&gt; = 5034), mostly heterosexual (&lt;i&gt;n&lt;/i&gt; = 1825), bisexual (&lt;i&gt;n&lt;/i&gt; = 394), and lesbian (&lt;i&gt;n&lt;/i&gt; = 996). These groups were compared to a “reference” group that self-identified as lifelong heterosexual (&lt;i&gt;n&lt;/i&gt; = 93,294). The researchers also determined the case numbers, incidence rates, and age-adjusted incidence rate ratios (aIRRs) of 21 site-specific cancers for each group. Using aIRRs, they compared incidence rates between the reference group and the four SO subgroups.&lt;/p&gt;&lt;p&gt;The researchers reported that the cancer incidence rate (cases per 100,000 person-years) was highest for those ","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":null,"pages":null},"PeriodicalIF":503.1,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.3322/caac.21861","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142118514","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Osimertinib prolongs progression-free lung cancer survival after chemotherapy 奥希替尼延长肺癌化疗后的无进展生存期
IF 503.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-02 DOI: 10.3322/caac.21862
Mike Fillon
<p>Investigators for the phase 3 LAURA trial reported that for patients with unresectable stage III non–small cell lung cancer (NSCLC) harboring an <i>EGFR</i> mutation after chemoradiotherapy, osimertinib significantly prolonged progression-free survival (PFS) versus a placebo. The study appears in <i>The New England Journal of Medicine</i> (doi:10.1056/NEJMoa2402614).</p><p>“This is the first randomized trial conducted specifically for patients with unresectable stage III lung cancer with <i>EGFR</i> mutation,” says Suresh S. Ramalingam, MD, professor of hematology and medical oncology and executive director of the Winship Cancer Institute at the Emory University School of Medicine and LAURA trial investigator. “The study shows a clear benefit with osimertinib compared to placebo in terms of progression-free survival and lower risk of brain progression.”</p><p>Study results show that treatment with osimertinib reduced the risk of progression or death by 84% in comparison with the placebo. The study was presented during a press briefing at the 2024 annual meeting of the American Society of Clinical Oncology held in Chicago, Illinois.</p><p>The LAURA trial included patients at least 18 years old (20 years old in Japan) with locally advanced and unresectable stage III NSCLC harboring an <i>EGFR</i> exon 19 deletion or L858R mutation from August 2018 through July 2022. None of the patients had disease progression after definitive chemoradiotherapy. A total of 216 patients underwent randomization; 143 were assigned to receive osimertinib, and 73 received the placebo.</p><p>The median age of the patients was 62 years in the osimertinib arm and 64 years in the placebo group. Most were male (63% in the osimertinib arm and 58% in the placebo arm). The majority never smoked (71% and 67%, respectively).</p><p>Both groups included enrollment from Asian countries (81% in the osimertinib arm vs. 85% in the placebo arm), had stage IIIB disease (47% vs. 52%) and adenocarcinoma (97% vs. 95%), and harbored <i>EGFR</i> exon 19 deletions (52% vs. 59%). Also, most received concurrent chemoradiotherapy: 92% in the osimertinib group and 85% in the placebo group.</p><p>Patients needed to have a World Health Organization (WHO) performance-status score of 0 or 1. (Note that this was based on a scale of 0–5, with a higher number indicating more disability.) Six weeks was the maximum interval between the last dose of chemoradiotherapy and randomization. At the baseline, patients were largely balanced between the two groups, although there were more patients with a WHO performance-status score of 0 in the osimertinib group versus the placebo group (56% vs. 42%).</p><p>The patients were randomly assigned 2:1 to receive 80 mg of oral osimertinib or the placebo once per day. Blinded independent central review (BICR) of PFS per the Response Evaluation Criteria in Solid Tumors (Version 1.1) was the trial’s primary end point. Secondary end points included overall survival (OS),
3期LAURA试验的研究人员报告说,对于化放疗后不可切除的携带表皮生长因子受体突变的III期非小细胞肺癌(NSCLC)患者,与安慰剂相比,奥希替尼能显著延长无进展生存期(PFS)。这项研究发表在《新英格兰医学杂志》上(doi:10.1056/NEJMoa2402614)。"这是第一项专门针对EGFR突变的不可切除III期肺癌患者进行的随机试验,"埃默里大学医学院血液学和肿瘤内科学教授、温希普癌症研究所执行主任、LAURA试验研究者Suresh S. Ramalingam医学博士说。"研究结果显示,与安慰剂相比,使用奥希替尼治疗可将病情恶化或死亡风险降低84%。这项研究是在伊利诺伊州芝加哥市举行的美国临床肿瘤学会2024年年会上的新闻发布会上公布的。"LAURA "试验纳入了2018年8月至2022年7月期间年满18岁(日本为20岁)、携带表皮生长因子受体外显子19缺失或L858R突变的局部晚期和不可切除的III期NSCLC患者。这些患者在接受明确的化放疗后均没有出现疾病进展。共有216名患者接受了随机分配,其中143人被分配接受奥希替尼治疗,73人接受安慰剂治疗。大多数患者为男性(奥希莫替尼组为63%,安慰剂组为58%)。两组患者均来自亚洲国家(奥西替尼组为81%,安慰剂组为85%),均为IIIB期疾病(47%对52%)和腺癌(97%对95%),均携带表皮生长因子受体外显子19缺失(52%对59%)。此外,大多数患者同时接受了化疗放疗:奥西美替尼组为92%,安慰剂组为85%。患者的世界卫生组织(WHO)表现状态评分需为0或1分(请注意,评分标准为0-5分,数字越大表示残疾程度越高)。最后一次化放疗剂量与随机化之间的最长间隔为六周。在基线时,两组患者的情况基本平衡,但奥希替尼组与安慰剂组相比,WHO表现状态评分为0分的患者更多(56%对42%)。患者按2:1的比例随机分配,每天一次口服80毫克奥希替尼或安慰剂。根据《实体瘤反应评价标准》(1.1版)进行的盲法独立中央审查(BICR)是该试验的主要终点。次要终点包括总生存期(OS)、中枢神经系统 PFS 和安全性。治疗一直持续到 BICR 确定的疾病进展、不可接受的毒性或事先商定的其他终止标准出现为止。肿瘤评估最初通过胸部CT/MRI和脑部MRI进行,第48周之前每8周评估一次,之后每12周评估一次,直到BICR确认病情进展。在两组患者中,只有8%的患者无法进行评估。奥西替尼组的中位应答持续时间为36.9个月,安慰剂组为6.5个月。其他数据显示,奥西替尼组有22名患者出现了新病灶,而安慰剂组有68名患者出现了新病灶。出现新病变的部位包括大脑(8 名奥希替尼患者对 29 名安慰剂患者)、肺部(6 名患者对 29 名患者)和肝脏(3 名患者对 7 名患者)。Ramalingam 博士说,化放疗后奥希替尼的研究结果符合 EGFR 抑制剂已知的安全性特征,而且副作用是可控的。他补充说,大多数不良反应都很轻微(1/2级),不会导致治疗中断,而且与之前的研究结果一致。至少有10%的患者出现了任何级别的不良反应,其中最常见的不良反应包括放射性肺炎(奥西替尼组48%的患者与安慰剂组38%的患者)。在至少10%的患者中,最常见的任何级别的不良反应包括放射性肺炎(奥希莫替尼组48%的患者对安慰剂组38%的患者)、腹泻(36%对14%)、皮疹(24%对14%)、COVID-19(20%对8%)、副癣(17%对1%)和咳嗽(16%对10%)。此外,奥希替尼组和安慰剂组还出现了食欲下降(15% vs. 5%)、皮肤干燥(13% vs. 5%)、瘙痒(13% vs. 7%)、口腔炎(12% vs. 3%)、白细胞计数下降(12% vs. 3%)、肺炎(11% vs. 8%)、贫血(10% vs. 4%)和肌肉骨骼胸痛(3% vs. 12%)。
{"title":"Osimertinib prolongs progression-free lung cancer survival after chemotherapy","authors":"Mike Fillon","doi":"10.3322/caac.21862","DOIUrl":"10.3322/caac.21862","url":null,"abstract":"&lt;p&gt;Investigators for the phase 3 LAURA trial reported that for patients with unresectable stage III non–small cell lung cancer (NSCLC) harboring an &lt;i&gt;EGFR&lt;/i&gt; mutation after chemoradiotherapy, osimertinib significantly prolonged progression-free survival (PFS) versus a placebo. The study appears in &lt;i&gt;The New England Journal of Medicine&lt;/i&gt; (doi:10.1056/NEJMoa2402614).&lt;/p&gt;&lt;p&gt;“This is the first randomized trial conducted specifically for patients with unresectable stage III lung cancer with &lt;i&gt;EGFR&lt;/i&gt; mutation,” says Suresh S. Ramalingam, MD, professor of hematology and medical oncology and executive director of the Winship Cancer Institute at the Emory University School of Medicine and LAURA trial investigator. “The study shows a clear benefit with osimertinib compared to placebo in terms of progression-free survival and lower risk of brain progression.”&lt;/p&gt;&lt;p&gt;Study results show that treatment with osimertinib reduced the risk of progression or death by 84% in comparison with the placebo. The study was presented during a press briefing at the 2024 annual meeting of the American Society of Clinical Oncology held in Chicago, Illinois.&lt;/p&gt;&lt;p&gt;The LAURA trial included patients at least 18 years old (20 years old in Japan) with locally advanced and unresectable stage III NSCLC harboring an &lt;i&gt;EGFR&lt;/i&gt; exon 19 deletion or L858R mutation from August 2018 through July 2022. None of the patients had disease progression after definitive chemoradiotherapy. A total of 216 patients underwent randomization; 143 were assigned to receive osimertinib, and 73 received the placebo.&lt;/p&gt;&lt;p&gt;The median age of the patients was 62 years in the osimertinib arm and 64 years in the placebo group. Most were male (63% in the osimertinib arm and 58% in the placebo arm). The majority never smoked (71% and 67%, respectively).&lt;/p&gt;&lt;p&gt;Both groups included enrollment from Asian countries (81% in the osimertinib arm vs. 85% in the placebo arm), had stage IIIB disease (47% vs. 52%) and adenocarcinoma (97% vs. 95%), and harbored &lt;i&gt;EGFR&lt;/i&gt; exon 19 deletions (52% vs. 59%). Also, most received concurrent chemoradiotherapy: 92% in the osimertinib group and 85% in the placebo group.&lt;/p&gt;&lt;p&gt;Patients needed to have a World Health Organization (WHO) performance-status score of 0 or 1. (Note that this was based on a scale of 0–5, with a higher number indicating more disability.) Six weeks was the maximum interval between the last dose of chemoradiotherapy and randomization. At the baseline, patients were largely balanced between the two groups, although there were more patients with a WHO performance-status score of 0 in the osimertinib group versus the placebo group (56% vs. 42%).&lt;/p&gt;&lt;p&gt;The patients were randomly assigned 2:1 to receive 80 mg of oral osimertinib or the placebo once per day. Blinded independent central review (BICR) of PFS per the Response Evaluation Criteria in Solid Tumors (Version 1.1) was the trial’s primary end point. Secondary end points included overall survival (OS), ","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":null,"pages":null},"PeriodicalIF":503.1,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.3322/caac.21862","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142118515","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Overlooked barriers to implementation of geriatric assessment 被忽视的实施老年评估的障碍
IF 254.7 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-29 DOI: 10.3322/caac.21865
Banu E. Symington, Paul G. Montgomery
<p>In this issue of the journal, Magnuson et al. provide a comprehensive review of available geriatric assessment (GA) tools and their impact on outcomes for solid tumors and hematologic malignancies. In addition, the authors provide a clear guide for clinicians to help understand the importance of GA and management.<span><sup>1</sup></span></p><p>An assumption inherent in the GA is that improvement in outcomes is driven by modifications in treatment delivery or implementation of features to make activities of daily living safer. In other words, GA-guided management<span><sup>2</sup></span> or GA-driven intervention,<span><sup>3</sup></span> rather than simply performing the GA, is what leads to outcome improvement. These modifications are implemented using a multidisciplinary team of geriatric trained specialists. Examples include occupational therapists to improve home safety, physical therapists to improve gait and balance, pharmacists to review home medications and adjust based on anticipated adverse drug interactions, dietitians to improve nutrition, etc. Most of the studies included showed benefit, either in survival, reduced toxicity, improved quality of life, or cost effectiveness.</p><p>These observations led to the development of an American Society of Clinical Oncology guideline recommending GA-guided management of cancer treatment in elderly adults.<span><sup>4</sup></span> However, it is widely recognized that this tool is underused by practicing oncologists.<span><sup>5</sup></span> The <i>whys</i> have been explored by Magnuson et al. and others<span><sup>5, 6</sup></span> and included the belief that the GA was too cumbersome in addition to the perception that it added little or no value. Based on these assumptions, making assessment tools more efficient and educating providers about their evidence-generated benefits have been the focus of efforts to improve GA use. To encourage greater uptake of the tool, Magnuson and co-authors detail ways to educate providers and simplify the GA.</p><p>What is not discussed that may be an important root cause of poor uptake of GA is resource scarcity, which takes two forms. The first is the lack of available services to support GA-modified treatment. Substantial numbers of communities, particularly in rural sites, do not have consistent—if any—access to the specialists required to modify treatment in a GA-guided manner. These practices almost certainly do not have geriatricians or geriatric-trained nurse practitioners; and they may not have physical therapists, occupational therapists, chemotherapy-dedicated pharmacists, or even social workers. Rural sites particularly often have one oncology provider whose job is to meet all the needs of every oncology patient in their practice. This distributive inequity of resources<span><sup>7</sup></span> has always existed and will continue to plague rural communities. In this context, even if one performed a GA, opportunities to make care delivery saf
远程医疗在肿瘤学的许多方面都取得了成功,从监测到毒性监测,并可用于老年病咨询和管理。我们的全国性组织(如美国临床肿瘤学会和美国癌症学会)可以帮助建立一个由全国认证的老年病学专家组成的小组,提供远程会诊,以解决老年病这方面的问题。不过,这要假定有足够多的老年病学专家愿意并能够满足日益增长的老年人口不断扩大的需求。一个更难解决的问题是,如何组建一个多学科团队来实现老年医学的建议,包括更广泛地获得职业和物理治疗的评估和管理,这需要亲临现场、亲力亲为的访问。解决这一人力问题的新方法或许是帮助创建一个新的辅助专业领域,即老年评估实施技术人员(GAITs)。这种老年评估实施技术员将由非专业人员担任,他们将接受相对短暂的集中培训,然后获得认证,类似于非专业导航员。这些人经过培训后,可以进行居家评估,为安全实施化疗做出必要的改变,并可部署到服务不足的社区和农村地区。如果设计得当,在缺乏经济机会的社区,这可能是一个有吸引力且有用的职业选择,并能为老龄化人口所代表的日益增长的需求提供解决方案。这两个要素,即支持当地医生的远程保健老年医学和补充远程保健职业治疗/物理治疗的 GAIT,将需要额外的资金,并需要实施研究,以确定这些增强措施是否和/或如何改善结果。设想支持这些举措所需的资源似乎令人生畏。如果我们想改善 GA 在所有肿瘤治疗实践中的应用,仅仅教育肿瘤学家和简化工具是不够的。我们必须帮助资源不足的诊疗机构实施基于 GA 的变革,我们需要将所需的专科诊疗带到农村环境中。牢记服务不足社区的需求对于继续推出的所有指南取得成功至关重要。
{"title":"Overlooked barriers to implementation of geriatric assessment","authors":"Banu E. Symington, Paul G. Montgomery","doi":"10.3322/caac.21865","DOIUrl":"https://doi.org/10.3322/caac.21865","url":null,"abstract":"&lt;p&gt;In this issue of the journal, Magnuson et al. provide a comprehensive review of available geriatric assessment (GA) tools and their impact on outcomes for solid tumors and hematologic malignancies. In addition, the authors provide a clear guide for clinicians to help understand the importance of GA and management.&lt;span&gt;&lt;sup&gt;1&lt;/sup&gt;&lt;/span&gt;&lt;/p&gt;\u0000&lt;p&gt;An assumption inherent in the GA is that improvement in outcomes is driven by modifications in treatment delivery or implementation of features to make activities of daily living safer. In other words, GA-guided management&lt;span&gt;&lt;sup&gt;2&lt;/sup&gt;&lt;/span&gt; or GA-driven intervention,&lt;span&gt;&lt;sup&gt;3&lt;/sup&gt;&lt;/span&gt; rather than simply performing the GA, is what leads to outcome improvement. These modifications are implemented using a multidisciplinary team of geriatric trained specialists. Examples include occupational therapists to improve home safety, physical therapists to improve gait and balance, pharmacists to review home medications and adjust based on anticipated adverse drug interactions, dietitians to improve nutrition, etc. Most of the studies included showed benefit, either in survival, reduced toxicity, improved quality of life, or cost effectiveness.&lt;/p&gt;\u0000&lt;p&gt;These observations led to the development of an American Society of Clinical Oncology guideline recommending GA-guided management of cancer treatment in elderly adults.&lt;span&gt;&lt;sup&gt;4&lt;/sup&gt;&lt;/span&gt; However, it is widely recognized that this tool is underused by practicing oncologists.&lt;span&gt;&lt;sup&gt;5&lt;/sup&gt;&lt;/span&gt; The &lt;i&gt;whys&lt;/i&gt; have been explored by Magnuson et al. and others&lt;span&gt;&lt;sup&gt;5, 6&lt;/sup&gt;&lt;/span&gt; and included the belief that the GA was too cumbersome in addition to the perception that it added little or no value. Based on these assumptions, making assessment tools more efficient and educating providers about their evidence-generated benefits have been the focus of efforts to improve GA use. To encourage greater uptake of the tool, Magnuson and co-authors detail ways to educate providers and simplify the GA.&lt;/p&gt;\u0000&lt;p&gt;What is not discussed that may be an important root cause of poor uptake of GA is resource scarcity, which takes two forms. The first is the lack of available services to support GA-modified treatment. Substantial numbers of communities, particularly in rural sites, do not have consistent—if any—access to the specialists required to modify treatment in a GA-guided manner. These practices almost certainly do not have geriatricians or geriatric-trained nurse practitioners; and they may not have physical therapists, occupational therapists, chemotherapy-dedicated pharmacists, or even social workers. Rural sites particularly often have one oncology provider whose job is to meet all the needs of every oncology patient in their practice. This distributive inequity of resources&lt;span&gt;&lt;sup&gt;7&lt;/sup&gt;&lt;/span&gt; has always existed and will continue to plague rural communities. In this context, even if one performed a GA, opportunities to make care delivery saf","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":null,"pages":null},"PeriodicalIF":254.7,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142100723","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Geriatric assessment for the practicing clinician: The why, what, and how 临床执业医师的老年病评估:为什么、做什么、怎么做
IF 254.7 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-29 DOI: 10.3322/caac.21864
Allison Magnuson, Kah Poh Loh, Fiona Stauffer, William Dale, Nikesha Gilmore, Sindhuja Kadambi, Heidi D. Klepin, Kaitlin Kyi, Lisa M. Lowenstein, Tanyanika Phillips, Erika Ramsdale, Melody K. Schiaffino, John F. Simmons, Grant R. Williams, Jason Zittel, Supriya Mohile
Older adults with cancer heterogeneously experience health care, treatment, and symptoms. Geriatric assessment (GA) offers a comprehensive evaluation of an older individual's health status and can predict cancer-related outcomes in individuals with solid tumors and those with hematologic malignancies. In the last decade, randomized controlled trials have demonstrated the benefits of GA and GA management (GAM), which uses GA information to provide tailored intervention strategies to address GA impairments (e.g., implementing physical therapy for impaired physical function). Multiple phase 3 clinical trials in older adults with solid tumors and hematologic malignancies have demonstrated that GAM improves treatment completion, quality of life, communication, and advance care planning while reducing treatment-related toxicity, falls, and polypharmacy. Nonetheless, implementation and uptake of GAM remain challenging. Various strategies have been proposed, including the use of GA screening tools, to identify patients most likely to benefit from GAM, the systematic engagement of the oncology workforce in the delivery of GAM, and the integration of technologies like telemedicine and mobile health to enhance the availability of GA and GAM interventions. Health inequities in minoritized groups persist, and systematic GA implementation has the potential to capture social determinants of health that are relevant to equitable care. Caregivers play an important role in cancer care and experience burden themselves. GA can guide dyadic supportive care interventions, ultimately helping both patients and caregivers achieve optimal health.
老年癌症患者在医疗保健、治疗和症状方面的经历各不相同。老年医学评估(GA)可对老年人的健康状况进行全面评估,并可预测实体瘤患者和血液恶性肿瘤患者的癌症相关预后。在过去的十年中,随机对照试验证明了老年医学评估和老年医学管理(GAM)的益处,老年医学管理利用老年医学评估信息提供量身定制的干预策略,以解决老年医学评估的缺陷(例如,针对受损的身体功能实施物理治疗)。在患有实体瘤和血液系统恶性肿瘤的老年人中开展的多项三期临床试验表明,GAM 可改善治疗完成度、生活质量、沟通和预先护理计划,同时减少治疗相关毒性、跌倒和多重用药。然而,GAM 的实施和普及仍面临挑战。人们提出了各种策略,包括使用GA筛查工具来识别最有可能从GAM中获益的患者,让肿瘤科工作人员系统地参与GAM的实施,以及整合远程医疗和移动医疗等技术来提高GA和GAM干预措施的可用性。少数群体的健康不平等现象依然存在,系统性地实施性别问题评估有可能捕捉到与公平护理相关的健康社会决定因素。护理人员在癌症护理中扮演着重要角色,他们自身也承受着负担。性别问题可以指导双向支持性护理干预,最终帮助患者和护理人员达到最佳健康状态。
{"title":"Geriatric assessment for the practicing clinician: The why, what, and how","authors":"Allison Magnuson, Kah Poh Loh, Fiona Stauffer, William Dale, Nikesha Gilmore, Sindhuja Kadambi, Heidi D. Klepin, Kaitlin Kyi, Lisa M. Lowenstein, Tanyanika Phillips, Erika Ramsdale, Melody K. Schiaffino, John F. Simmons, Grant R. Williams, Jason Zittel, Supriya Mohile","doi":"10.3322/caac.21864","DOIUrl":"https://doi.org/10.3322/caac.21864","url":null,"abstract":"Older adults with cancer heterogeneously experience health care, treatment, and symptoms. Geriatric assessment (GA) offers a comprehensive evaluation of an older individual's health status and can predict cancer-related outcomes in individuals with solid tumors and those with hematologic malignancies. In the last decade, randomized controlled trials have demonstrated the benefits of GA and GA management (GAM), which uses GA information to provide tailored intervention strategies to address GA impairments (e.g., implementing physical therapy for impaired physical function). Multiple phase 3 clinical trials in older adults with solid tumors and hematologic malignancies have demonstrated that GAM improves treatment completion, quality of life, communication, and advance care planning while reducing treatment-related toxicity, falls, and polypharmacy. Nonetheless, implementation and uptake of GAM remain challenging. Various strategies have been proposed, including the use of GA screening tools, to identify patients most likely to benefit from GAM, the systematic engagement of the oncology workforce in the delivery of GAM, and the integration of technologies like telemedicine and mobile health to enhance the availability of GA and GAM interventions. Health inequities in minoritized groups persist, and systematic GA implementation has the potential to capture social determinants of health that are relevant to equitable care. Caregivers play an important role in cancer care and experience burden themselves. GA can guide dyadic supportive care interventions, ultimately helping both patients and caregivers achieve optimal health.","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":null,"pages":null},"PeriodicalIF":254.7,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142100971","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Proportion and number of cancer cases and deaths attributable to potentially modifiable risk factors in the United States, 2019 2019 年美国可归因于潜在可改变风险因素的癌症病例和死亡病例的比例和数量。
IF 503.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-11 DOI: 10.3322/caac.21858
Farhad Islami MD, PhD, Emily C. Marlow PhD, Blake Thomson DPhil, MPhil, Marjorie L. McCullough ScD, RD, Harriet Rumgay PhD, Susan M. Gapstur PhD, MPH, Alpa V. Patel PhD, Isabelle Soerjomataram MD, PhD, MSc, Ahmedin Jemal DVM, PhD

In 2018, the authors reported estimates of the number and proportion of cancers attributable to potentially modifiable risk factors in 2014 in the United States. These data are useful for advocating for and informing cancer prevention and control. Herein, based on up-to-date relative risk and cancer occurrence data, the authors estimated the proportion and number of invasive cancer cases (excluding nonmelanoma skin cancers) and deaths, overall and for 30 cancer types among adults who were aged 30 years and older in 2019 in the United States, that were attributable to potentially modifiable risk factors. These included cigarette smoking; second-hand smoke; excess body weight; alcohol consumption; consumption of red and processed meat; low consumption of fruits and vegetables, dietary fiber, and dietary calcium; physical inactivity; ultraviolet radiation; and seven carcinogenic infections. Numbers of cancer cases and deaths were obtained from data sources with complete national coverage, risk factor prevalence estimates from nationally representative surveys, and associated relative risks of cancer from published large-scale pooled or meta-analyses. In 2019, an estimated 40.0% (713,340 of 1,781,649) of all incident cancers (excluding nonmelanoma skin cancers) and 44.0% (262,120 of 595,737) of all cancer deaths in adults aged 30 years and older in the United States were attributable to the evaluated risk factors. Cigarette smoking was the leading risk factor contributing to cancer cases and deaths overall (19.3% and 28.5%, respectively), followed by excess body weight (7.6% and 7.3%, respectively), and alcohol consumption (5.4% and 4.1%, respectively). For 19 of 30 evaluated cancer types, more than one half of the cancer cases and deaths were attributable to the potentially modifiable risk factors considered in this study. Lung cancer had the highest number of cancer cases (201,660) and deaths (122,740) attributable to evaluated risk factors, followed by female breast cancer (83,840 cases), skin melanoma (82,710), and colorectal cancer (78,440) for attributable cases and by colorectal (25,800 deaths), liver (14,720), and esophageal (13,600) cancer for attributable deaths. Large numbers of cancer cases and deaths in the United States are attributable to potentially modifiable risk factors, underscoring the potential to substantially reduce the cancer burden through broad and equitable implementation of preventive initiatives.

2018 年,作者报告了美国 2014 年可归因于潜在可改变风险因素的癌症数量和比例的估计值。这些数据有助于倡导癌症预防和控制并为其提供信息。在此,作者根据最新的相对风险和癌症发生率数据,估算了2019年美国30岁及以上成年人中,可归因于潜在可改变风险因素的浸润性癌症病例(不包括非黑色素瘤皮肤癌)和死亡病例的比例和数量。这些因素包括吸烟、二手烟、体重超标、饮酒、食用红肉和加工肉类、水果和蔬菜、膳食纤维和膳食钙摄入量低、缺乏运动、紫外线辐射以及七种致癌感染。癌症病例和死亡人数来自覆盖全国的数据源、全国代表性调查得出的风险因素流行率估计值,以及已发表的大规模汇总分析或荟萃分析得出的相关癌症相对风险系数。2019年,在美国30岁及以上成年人中,估计有40.0%(1,781,649人中的713,340人)的癌症发病率(不包括非黑色素瘤皮肤癌)和44.0%(595,737人中的262,120人)的癌症死亡率可归因于所评估的风险因素。吸烟是导致癌症病例和死亡的首要风险因素(分别为 19.3% 和 28.5%),其次是超重(分别为 7.6% 和 7.3%)和饮酒(分别为 5.4% 和 4.1%)。在 30 种被评估的癌症类型中,有 19 种癌症的病例和死亡人数的一半以上可归因于本研究中考虑的潜在可改变风险因素。肺癌的病例数(201,660 例)和死亡数(122,740 例)最高,其次是女性乳腺癌(83,840 例)、皮肤黑色素瘤(82,710 例)和结直肠癌(78,440 例),死亡数依次为结直肠癌(25,800 例死亡)、肝癌(14,720 例)和食道癌(13,600 例)。在美国,大量癌症病例和死亡病例可归因于潜在的可改变风险因素,这凸显了通过广泛而公平地实施预防措施来大幅减轻癌症负担的潜力。
{"title":"Proportion and number of cancer cases and deaths attributable to potentially modifiable risk factors in the United States, 2019","authors":"Farhad Islami MD, PhD,&nbsp;Emily C. Marlow PhD,&nbsp;Blake Thomson DPhil, MPhil,&nbsp;Marjorie L. McCullough ScD, RD,&nbsp;Harriet Rumgay PhD,&nbsp;Susan M. Gapstur PhD, MPH,&nbsp;Alpa V. Patel PhD,&nbsp;Isabelle Soerjomataram MD, PhD, MSc,&nbsp;Ahmedin Jemal DVM, PhD","doi":"10.3322/caac.21858","DOIUrl":"10.3322/caac.21858","url":null,"abstract":"<p>In 2018, the authors reported estimates of the number and proportion of cancers attributable to potentially modifiable risk factors in 2014 in the United States. These data are useful for advocating for and informing cancer prevention and control. Herein, based on up-to-date relative risk and cancer occurrence data, the authors estimated the proportion and number of invasive cancer cases (excluding nonmelanoma skin cancers) and deaths, overall and for 30 cancer types among adults who were aged 30 years and older in 2019 in the United States, that were attributable to potentially modifiable risk factors. These included cigarette smoking; second-hand smoke; excess body weight; alcohol consumption; consumption of red and processed meat; low consumption of fruits and vegetables, dietary fiber, and dietary calcium; physical inactivity; ultraviolet radiation; and seven carcinogenic infections. Numbers of cancer cases and deaths were obtained from data sources with complete national coverage, risk factor prevalence estimates from nationally representative surveys, and associated relative risks of cancer from published large-scale pooled or meta-analyses. In 2019, an estimated 40.0% (713,340 of 1,781,649) of all incident cancers (excluding nonmelanoma skin cancers) and 44.0% (262,120 of 595,737) of all cancer deaths in adults aged 30 years and older in the United States were attributable to the evaluated risk factors. Cigarette smoking was the leading risk factor contributing to cancer cases and deaths overall (19.3% and 28.5%, respectively), followed by excess body weight (7.6% and 7.3%, respectively), and alcohol consumption (5.4% and 4.1%, respectively). For 19 of 30 evaluated cancer types, more than one half of the cancer cases and deaths were attributable to the potentially modifiable risk factors considered in this study. Lung cancer had the highest number of cancer cases (201,660) and deaths (122,740) attributable to evaluated risk factors, followed by female breast cancer (83,840 cases), skin melanoma (82,710), and colorectal cancer (78,440) for attributable cases and by colorectal (25,800 deaths), liver (14,720), and esophageal (13,600) cancer for attributable deaths. Large numbers of cancer cases and deaths in the United States are attributable to potentially modifiable risk factors, underscoring the potential to substantially reduce the cancer burden through broad and equitable implementation of preventive initiatives.</p>","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":null,"pages":null},"PeriodicalIF":503.1,"publicationDate":"2024-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.3322/caac.21858","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141578363","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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 issues face AI deployment in cancer care 在癌症护理中应用人工智能面临的关键问题。
IF 503.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-04 DOI: 10.3322/caac.21860
Mike Fillon

With artificial intelligence (AI) erupting across all aspects of life, including health care, oncology is a logical field ripe for new applications. AI is already used in cancer care and diagnosis, such as tumor identification on x-rays and pathology slides. Beyond that, emerging technology is using AI to forecast the prognosis of patients and to assess their treatment options. One unknown is how oncologists feel about this trend, which includes possibly relinquishing some control over their profession and patients.

A new study asked 204 oncologists for their views on the rapidly developing AI tools. Specifically, they were asked about ethical issues that they face regarding the deployment of AI (e.g., whether they believed that AI could be used effectively in patient-care decisions). The main issue that the researchers investigated was to what degree patients should provide explicit informed consent for the use of AI during treatment decision-making. The study appears in JAMA Network Open (doi:10.1001/jamanetworkopen.2024.4077).

In the study, which was conducted from November 15, 2022 to July 31, 2023, a random sample of oncologists from across the country were asked 24 questions via traditional mail (which included a $25 gift card) about their views on the use of AI in clinical practice. Follow-ups with nonresponders were conducted via email and phone calls.

Issues covered bias, responsibilities, and whether they would be able to explain to patients how the technology was deployed in determining their care. There were 387 surveys sent to oncologists; 52.7% (n = 204) were completed. Those responding came from 37 states; 63.7% (n = 120) were male, and 62.7% (n = 128) identified as non-Hispanic White.

Very few respondents said that AI prognostic and clinical decision models could be used clinically when only researchers could explain them (13.2% of respondents [n = 27] for prognosis and 7.8% [n = 16] for clinical decisions).

For AI prognostic and clinical decision models that oncologists could explain, the percentages were much higher: 81.3% (n = 165) and 84.8% (n = 173), respectively. Fewer respondents—13.8% (n = 28) and 23.0% (n = 47), respectively—reported that the models also needed to be explainable by patients.

The survey also found that 36.8% of oncologists (n = 75) believed that if an AI system selected a treatment regimen different from what they would recommend, they would present both options and let the patient decide. Although that represented less than half of the respondents, it was the most common answer.

Regarding responsibility for medical or legal problems arising from AI use, 90.7% of respondents (n = 185) indicated that AI developers should be held accountable. This was considerably higher than the 47.1% (n = 96) who felt that the responsibility should be shared with physicians and the 43.1% (

随着人工智能(AI)在包括医疗保健在内的生活各个方面的爆发,肿瘤学也顺理成章地成为了一个成熟的新应用领域。人工智能已被用于癌症护理和诊断,如在 X 光片和病理切片上识别肿瘤。除此之外,新兴技术正在利用人工智能预测病人的预后并评估他们的治疗方案。一项新的研究询问了 204 名肿瘤学家对快速发展的人工智能工具的看法。具体来说,他们被问及在部署人工智能方面所面临的伦理问题(例如,他们是否认为人工智能可以有效地用于患者护理决策)。研究人员调查的主要问题是,在治疗决策过程中使用人工智能时,患者应在多大程度上提供明确的知情同意。这项研究发表在《美国医学会杂志网络版》(JAMA Network Open)上(doi:10.1001/jamanetworkopen.2024.4077)。研究于2022年11月15日至2023年7月31日进行,研究人员通过传统邮件(其中包括一张价值25美元的礼品卡)随机抽取了全国各地的肿瘤学家,向他们提出了24个问题,询问他们对在临床实践中使用人工智能的看法。问题涉及偏见、责任以及他们是否能够向患者解释在决定他们的治疗时是如何部署该技术的。共向肿瘤学家发送了 387 份调查问卷;52.7%(n = 204)的人完成了问卷。受访者来自美国 37 个州;63.7%(n=120)为男性,62.7%(n=128)为非西班牙裔白人。对于肿瘤学家可以解释的人工智能预后和临床决策模型,百分比要高得多:分别为 81.3%(n = 165)和 84.8%(n = 173)。调查还发现,36.8% 的肿瘤学家(n = 75)认为,如果人工智能系统选择的治疗方案与他们推荐的不同,他们会提出两种方案,让患者决定。关于人工智能使用过程中产生的医疗或法律问题的责任,90.7% 的受访者(n = 185)表示人工智能开发者应承担责任。尽管76.5%的受访者(n = 156)指出,肿瘤学家应保护患者免受有偏见的人工智能工具(如:使用的非通用数据集)的影响,但这一比例远高于认为应由医生分担责任的47.1%(n = 96)和认为应由医院分担责任的43.1%(n = 88)、尽管 76.5%的受访者(n = 156)指出,肿瘤学家应保护患者免受有偏见的人工智能工具(例如,用于为患者提供护理信息的非通用数据集)的影响,但只有 27.9%(n = 57)的受访者认为他们能够识别出反映偏见的人工智能模型。"这项研究非常重要,"德克萨斯肿瘤医院的肿瘤内科医生、第一阶段研究员、德克萨斯州达拉斯市 NEXT 肿瘤医院临床研究主任 Shiraj Sen 博士说。他认为,该技术的发展速度远远超过了临床医生对其影响的认识。"虽然肿瘤学领域的人工智能工具正在迅速发展,但很少有研究能捕捉到肿瘤学家对谁将负责其使用的伦理领域的观点。"森博士补充说,"现在是肿瘤学家开始思考和讨论其中细微差别的时候了。这项研究有助于凸显许多肿瘤学家已经开始出现的意见分歧,并强调有必要作为一个群体就使用人工智能辅助工具时肿瘤学家和患者之间如何分担决策责任进行更广泛的讨论。"该研究的作者安德鲁-汉特尔(Andrew Hantel)医学博士是哈佛大学医学院的医学讲师,同时也是马萨诸塞州波士顿市丹娜-法伯癌症研究所白血病和人口科学部门以及哈佛大学医学院生命伦理学中心的教员。他指出,随着人工智能开始影响癌症护理的提供,了解那些将被要求实施人工智能的人--肿瘤学家--的伦理影响至关重要。Hantel 博士补充说,这项调查旨在为这一领域提供数据,并重点关注可解释性、同意、责任和公平等伦理问题。 "我们的目的是提出执业肿瘤学家的观点,以便以符合伦理的方式部署人工智能,满足肿瘤学家和患者的需求,同时解决潜在的伦理困境。"汉特尔博士说,在这项调查之前,利益相关者对这些伦理问题的看法并不为人所知。他补充说,除了新颖性之外,这项研究的重要性还在于他们发现肿瘤学家在几个方面达成了共识:肿瘤学家有必要对人工智能模型进行解释,患者同意人工智能用于治疗决策的重要性,以及肿瘤学家坚信他们的专业职责包括保护患者免受有偏见的人工智能的伤害。"令人惊讶的是,相当多的受访者表示对识别人工智能模型中的偏见缺乏信心。这些观点的一致性突出表明,肿瘤学领域迫切需要结构化的人工智能教育和伦理准则。他补充说:"有趣的是,虽然肿瘤学家认为患者不需要能够解释人工智能模型,但当我们向他们提出人工智能不同意其治疗建议的情景时,最常见的反应是向患者提出两种选择,让他们自己决定。这一发现突出表明,许多医生不知道如何在人工智能方面采取行动,并就这种情况向患者提供咨询。"森博士认为,人工智能工具主要朝着三个方向发展。首先是治疗决策。"对于患者来说,幸运的是,新型治疗方案的出现为肿瘤医生提供了多种治疗选择,在特定的治疗环境下,任何一个患者都可以选择多种治疗方案。然而,这些治疗方案往往没有经过深入研究。能够帮助纳入预后因素、各种生物标志物和其他患者相关因素的人工智能工具可能很快就能在这种情况下提供帮助。"二是放射学反应评估。"使用人工智能辅助工具对抗癌治疗进行放射反应评估的临床试验已经在进行中。未来有一天,这些工具甚至可能帮助描述肿瘤异质性、预测治疗反应、评估肿瘤侵袭性,并帮助指导个性化治疗策略。"森博士说,最后一个领域是临床试验识别和评估。"每20个癌症患者中只有不到1人会参加临床试验。人工智能工具可能很快就能帮助确定适合患者的临床试验,甚至协助肿瘤学家初步评估患者有资格参加哪些试验。这些工具将有助于简化晚期癌症患者及其肿瘤学家获得临床试验的途径。"Sen 博士说,这些应用的准确性自然会存在隐患和问题。"在这些工具的开发过程中,肿瘤学家的广泛验证和密切参与可能有助于减少这些担忧。在人工智能这个话题上,我的建议是,随着人工智能工具的发展,所有肿瘤学家都要保持对人工智能工具的了解。正如我们从纸质病历过渡到 EMR(电子病历)时的情况一样,如果应用得当,有意识地使用人工智能工具可以帮助肿瘤学家高效、有效地提供优质医疗服务。"Hantel 博士说,要在肿瘤学领域合乎道德地部署人工智能,当务之急必须是开发支持肿瘤学家培训以及透明度、同意、问责和公平的基础设施。"这意味着需要围绕癌症人工智能开发基础设施,以确保其伦理部署。"Hantel 博士继续说,调查还发现了另一个必须认真对待的要点:需要了解患者--特别是那些历史上被边缘化和代表性不足的群体--对这些相同问题的看法。"然后,我们需要开发和测试用于开发和部署人工智能的伦理基础设施的有效性,以实现利益最大化和伤害最小化以及这些其他伦理问题,并对临床医生进行人工智能模型及其使用伦理方面的教育。"
{"title":"Key issues face AI deployment in cancer care","authors":"Mike Fillon","doi":"10.3322/caac.21860","DOIUrl":"10.3322/caac.21860","url":null,"abstract":"<p>With artificial intelligence (AI) erupting across all aspects of life, including health care, oncology is a logical field ripe for new applications. AI is already used in cancer care and diagnosis, such as tumor identification on x-rays and pathology slides. Beyond that, emerging technology is using AI to forecast the prognosis of patients and to assess their treatment options. One unknown is how oncologists feel about this trend, which includes possibly relinquishing some control over their profession and patients.</p><p>A new study asked 204 oncologists for their views on the rapidly developing AI tools. Specifically, they were asked about ethical issues that they face regarding the deployment of AI (e.g., whether they believed that AI could be used effectively in patient-care decisions). The main issue that the researchers investigated was to what degree patients should provide explicit informed consent for the use of AI during treatment decision-making. The study appears in <i>JAMA Network Open</i> (doi:10.1001/jamanetworkopen.2024.4077).</p><p>In the study, which was conducted from November 15, 2022 to July 31, 2023, a random sample of oncologists from across the country were asked 24 questions via traditional mail (which included a $25 gift card) about their views on the use of AI in clinical practice. Follow-ups with nonresponders were conducted via email and phone calls.</p><p>Issues covered bias, responsibilities, and whether they would be able to explain to patients how the technology was deployed in determining their care. There were 387 surveys sent to oncologists; 52.7% (<i>n</i> = 204) were completed. Those responding came from 37 states; 63.7% (<i>n</i> = 120) were male, and 62.7% (<i>n</i> = 128) identified as non-Hispanic White.</p><p>Very few respondents said that AI prognostic and clinical decision models could be used clinically when only researchers could explain them (13.2% of respondents [<i>n</i> = 27] for prognosis and 7.8% [<i>n</i> = 16] for clinical decisions).</p><p>For AI prognostic and clinical decision models that oncologists could explain, the percentages were much higher: 81.3% (<i>n</i> = 165) and 84.8% (<i>n</i> = 173), respectively. Fewer respondents—13.8% (<i>n</i> = 28) and 23.0% (<i>n</i> = 47), respectively—reported that the models also needed to be explainable by patients.</p><p>The survey also found that 36.8% of oncologists (<i>n</i> = 75) believed that if an AI system selected a treatment regimen different from what they would recommend, they would present both options and let the patient decide. Although that represented less than half of the respondents, it was the most common answer.</p><p>Regarding responsibility for medical or legal problems arising from AI use, 90.7% of respondents (<i>n</i> = 185) indicated that AI developers should be held accountable. This was considerably higher than the 47.1% (<i>n</i> = 96) who felt that the responsibility should be shared with physicians and the 43.1% (","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":null,"pages":null},"PeriodicalIF":503.1,"publicationDate":"2024-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.3322/caac.21860","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141532957","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Colon cancer blood test effective for average-risk population 结肠癌血液检测对一般风险人群有效。
IF 503.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-04 DOI: 10.3322/caac.21859
Mike Fillon

A new study appearing in The New England Journal of Medicine (NEJM) investigated the effectiveness of an emerging colon cancer screening option—a cell-free DNA (cfDNA) blood-based test known as Shield—that researchers and clinicians hope will encourage more people to be screened for colorectal cancer (CRC) (doi:10.1056/NEJMoa2304714).

Allison Rosen, MS, from Houston, Texas, is a 12-year CRC survivor who says that she is alive today because of timely colon cancer screening. “Unfortunately,” says Ms Rosen, “after talking with the community about the importance of screening and even with the knowledge that screening can save their life, people tell me every day that they refuse to get screened because both stool-based tests and colonoscopies have very negative stigmas.”

Ms Rosen points to American Cancer Society (ACS) data showing that one third of the screening-eligible population is not getting screened even though 90% of CRC deaths can be prevented with timely screening. Ms Rosen is the director of the ACS’s Project ECHO (Extension for Community Healthcare Outcomes) in Houston, Texas.

The Shield test is a blood-based CRC screening test meant for average-risk people with no family history of CRC and no personal history of CRC or advanced polyps (large polyps). Targeted patients are also CRC symptom–free.

The researchers who conducted the NEJM study believe that this screening test option could be key to improving screening rates, leading to fewer colon cancer deaths.

According to study author William M. Grady, MD, medical director of the Gastrointestinal Cancer Prevention Program at the Fred Hutchinson Cancer Center in Seattle, Washington, the test is on par with stool-based CRC screening tests, such as the fecal immunochemical test (FIT) and the Cologuard multitarget stool DNA test, for the detection of CRC.

The results of the NEJM study were derived from the ECLIPSE (Evaluation of the ctDNA LUNAR Test in an Average Patient Screening Episode) study, a multisite clinical trial of nearly 8000 people aged 45–84 years. The study was underwritten and led by Guardant Health (based in Palo Alto, California). The focus of the study was the comparison of the effectiveness of the blood test and colonoscopies for CRC sensitivity and for advanced neoplasia, including CRC and advanced precancerous lesions. Also investigated was the sensitivity for discovering if advanced precancerous lesions had a negative cfDNA blood-based test.

Of the 7861 patients who met the study criteria, 83.1% with colonoscopy-detected CRC registered a positive cfDNA test; 16.9% had a negative test (i.e., a colonoscopy detected CRC, but a circulating tumor DNA [ctDNA] test did not). Overall, the researchers found that the blood test was most sensitive for CRCs, including early-stage cancers. They also found that it was less sensitive for advanced precancerous lesions, which may become cancerous later.

Furthe

"奈斯医生说:"这是一种结肠癌检测试验,但对结肠癌的预防没有用处。"不过,我们还是希望有更多的人能因这项检测而接受筛查。"奈斯博士的另一个担忧是,如果循环癌DNA检测结果呈阳性,但后续的结肠镜检查却没有发现癌源,那该怎么办?"那么问题就来了,循环 DNA 从何而来?患者是否要无谓地担心,如果检测结果呈阳性,会增加身体其他部位患癌的风险,从而导致不必要的额外检查?"Grady 博士说,血液检测已经是 CRC 筛查的一种选择,当有人决定使用哪种 CRC 筛查检测时,可以与主治医生讨论。格雷迪博士说:"这一点意义重大,因为目前当人们可以选择用粪便检验或结肠镜检查来进行 CRC 筛查时,大约有三分之一的人选择两者都不做。"关于 ECLIPSE 试验和 "盾牌 "检验,还有两件事值得注意。首先,参加试验的人反映了美国总人口的人口统计学特征,包括按比例参加试验的非裔美国人/黑人、西班牙裔美国人和亚裔美国人。"Grady博士说,第二个关键点是Shield检测中使用的技术(ctDNA甲基组和片段组模式分析)在筛查检测中是新颖的。"对于基于血液的癌症筛查检验来说,这是一项非常有前景的技术,也是正在研究的其他基于血液的癌症筛查检验的基础,这些检验可用于筛查乳腺癌、肺癌和其他类型的癌症。"筛查倡导者罗森女士认为,与基于粪便的检验或结肠镜检查相比,在初级保健医生就诊时就可进行的血液检验可能会被证明是首选,而且依从性也会增加。"我相信,最好的筛查检测就是能做的检测。罗森女士补充说:"作为一名倡导者,我每天都在努力打破存在的污名,并强调筛查可以挽救生命。社区需要明白,他们需要在出现症状之前接受筛查,因为当他们开始出现症状时,被诊断为癌症晚期的可能性已经很高了。这项检查的目的是预防。我有许多朋友死于结肠直肠癌,因此让更多人接受筛查是我的一个目标。我想避免有人经历我所经历的一切"。
{"title":"Colon cancer blood test effective for average-risk population","authors":"Mike Fillon","doi":"10.3322/caac.21859","DOIUrl":"10.3322/caac.21859","url":null,"abstract":"<p>A new study appearing in The <i>New England Journal of Medicine</i> (<i>NEJM</i>) investigated the effectiveness of an emerging colon cancer screening option—a cell-free DNA (cfDNA) blood-based test known as Shield—that researchers and clinicians hope will encourage more people to be screened for colorectal cancer (CRC) (doi:10.1056/NEJMoa2304714).</p><p>Allison Rosen, MS, from Houston, Texas, is a 12-year CRC survivor who says that she is alive today because of timely colon cancer screening. “Unfortunately,” says Ms Rosen, “after talking with the community about the importance of screening and even with the knowledge that screening can save their life, people tell me every day that they refuse to get screened because both stool-based tests and colonoscopies have very negative stigmas.”</p><p>Ms Rosen points to American Cancer Society (ACS) data showing that one third of the screening-eligible population is not getting screened even though 90% of CRC deaths can be prevented with timely screening. Ms Rosen is the director of the ACS’s Project ECHO (Extension for Community Healthcare Outcomes) in Houston, Texas.</p><p>The Shield test is a blood-based CRC screening test meant for average-risk people with no family history of CRC and no personal history of CRC or advanced polyps (large polyps). Targeted patients are also CRC symptom–free.</p><p>The researchers who conducted the <i>NEJM</i> study believe that this screening test option could be key to improving screening rates, leading to fewer colon cancer deaths.</p><p>According to study author William M. Grady, MD, medical director of the Gastrointestinal Cancer Prevention Program at the Fred Hutchinson Cancer Center in Seattle, Washington, the test is on par with stool-based CRC screening tests, such as the fecal immunochemical test (FIT) and the Cologuard multitarget stool DNA test, for the detection of CRC.</p><p>The results of the <i>NEJM</i> study were derived from the ECLIPSE (Evaluation of the ctDNA LUNAR Test in an Average Patient Screening Episode) study, a multisite clinical trial of nearly 8000 people aged 45–84 years. The study was underwritten and led by Guardant Health (based in Palo Alto, California). The focus of the study was the comparison of the effectiveness of the blood test and colonoscopies for CRC sensitivity and for advanced neoplasia, including CRC and advanced precancerous lesions. Also investigated was the sensitivity for discovering if advanced precancerous lesions had a negative cfDNA blood-based test.</p><p>Of the 7861 patients who met the study criteria, 83.1% with colonoscopy-detected CRC registered a positive cfDNA test; 16.9% had a negative test (i.e., a colonoscopy detected CRC, but a circulating tumor DNA [ctDNA] test did not). Overall, the researchers found that the blood test was most sensitive for CRCs, including early-stage cancers. They also found that it was less sensitive for advanced precancerous lesions, which may become cancerous later.</p><p>Furthe","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":null,"pages":null},"PeriodicalIF":503.1,"publicationDate":"2024-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.3322/caac.21859","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141532956","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Cancer misinformation on social media 社交媒体上的癌症误导
IF 503.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-06-19 DOI: 10.3322/caac.21857
Stacy Loeb MD, MSc, PhD (Hon), Aisha T. Langford PhD, MPH, Marie A. Bragg PhD, Robert Sherman BA, June M. Chan ScD

Social media is widely used globally by patients, families of patients, health professionals, scientists, and other stakeholders who seek and share information related to cancer. Despite many benefits of social media for cancer care and research, there is also a substantial risk of exposure to misinformation, or inaccurate information about cancer. Types of misinformation vary from inaccurate information about cancer risk factors or unproven treatment options to conspiracy theories and public relations articles or advertisements appearing as reliable medical content. Many characteristics of social media networks—such as their extensive use and the relative ease it allows to share information quickly—facilitate the spread of misinformation. Research shows that inaccurate and misleading health-related posts on social media often get more views and engagement (e.g., likes, shares) from users compared with accurate information. Exposure to misinformation can have downstream implications for health-related attitudes and behaviors. However, combatting misinformation is a complex process that requires engagement from media platforms, scientific and health experts, governmental organizations, and the general public. Cancer experts, for example, should actively combat misinformation in real time and should disseminate evidence-based content on social media. Health professionals should give information prescriptions to patients and families and support health literacy. Patients and families should vet the quality of cancer information before acting upon it (e.g., by using publicly available checklists) and seek recommended resources from health care providers and trusted organizations. Future multidisciplinary research is needed to identify optimal ways of building resilience and combating misinformation across social media.

在全球范围内,患者、患者家属、医疗专业人员、科学家和其他利益相关者广泛使用社交媒体,寻求和分享与癌症有关的信息。尽管社交媒体对癌症护理和研究有很多益处,但也存在接触错误信息或不准确癌症信息的巨大风险。错误信息的类型多种多样,有关于癌症风险因素或未经证实的治疗方案的不准确信息,也有阴谋论、公关文章或广告冒充可靠的医疗内容。社交媒体网络的许多特点--如其广泛的使用和相对容易的快速信息共享--为错误信息的传播提供了便利。研究表明,与准确的信息相比,社交媒体上与健康相关的不准确和误导性帖子往往能获得更多用户的浏览和参与(如点赞、分享)。接触错误信息会对健康相关的态度和行为产生下游影响。然而,打击错误信息是一个复杂的过程,需要媒体平台、科学和健康专家、政府组织以及公众的参与。例如,癌症专家应实时积极地与错误信息作斗争,并在社交媒体上传播以证据为基础的内容。医疗专业人员应向患者和家属提供信息处方,支持健康知识普及。患者和家属在根据癌症信息采取行动之前,应审查信息的质量(例如,通过使用公开可用的核对表),并向医疗服务提供者和可信赖的组织寻求推荐资源。未来需要开展多学科研究,以确定建立复原力和打击社交媒体错误信息的最佳方法。
{"title":"Cancer misinformation on social media","authors":"Stacy Loeb MD, MSc, PhD (Hon),&nbsp;Aisha T. Langford PhD, MPH,&nbsp;Marie A. Bragg PhD,&nbsp;Robert Sherman BA,&nbsp;June M. Chan ScD","doi":"10.3322/caac.21857","DOIUrl":"10.3322/caac.21857","url":null,"abstract":"<p>Social media is widely used globally by patients, families of patients, health professionals, scientists, and other stakeholders who seek and share information related to cancer. Despite many benefits of social media for cancer care and research, there is also a substantial risk of exposure to misinformation, or inaccurate information about cancer. Types of misinformation vary from inaccurate information about cancer risk factors or unproven treatment options to conspiracy theories and public relations articles or advertisements appearing as reliable medical content. Many characteristics of social media networks—such as their extensive use and the relative ease it allows to share information quickly—facilitate the spread of misinformation. Research shows that inaccurate and misleading health-related posts on social media often get more views and engagement (e.g., likes, shares) from users compared with accurate information. Exposure to misinformation can have downstream implications for health-related attitudes and behaviors. However, combatting misinformation is a complex process that requires engagement from media platforms, scientific and health experts, governmental organizations, and the general public. Cancer experts, for example, should actively combat misinformation in real time and should disseminate evidence-based content on social media. Health professionals should give <i>information prescriptions</i> to patients and families and support health literacy. Patients and families should vet the quality of cancer information before acting upon it (e.g., by using publicly available checklists) and seek recommended resources from health care providers and trusted organizations. Future multidisciplinary research is needed to identify optimal ways of building resilience and combating misinformation across social media.</p>","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":null,"pages":null},"PeriodicalIF":503.1,"publicationDate":"2024-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.3322/caac.21857","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141425293","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
The evolving landscape of tissue-agnostic therapies in precision oncology 精准肿瘤学中组织诊断疗法不断发展的前景。
IF 503.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-05-30 DOI: 10.3322/caac.21844
Vivek Subbiah MD, Mohamed A. Gouda MD, Bettina Ryll MD, PhD, Howard A. Burris III MD, Razelle Kurzrock MD

Tumor-agnostic therapies represent a paradigm shift in oncology by altering the traditional means of characterizing tumors based on their origin or location. Instead, they zero in on specific genetic anomalies responsible for fueling malignant growth. The watershed moment for tumor-agnostic therapies arrived in 2017, with the US Food and Drug Administration's historic approval of pembrolizumab, an immune checkpoint inhibitor. This milestone marked the marriage of genomics and immunology fields, as an immunotherapeutic agent gained approval based on genomic biomarkers, specifically, microsatellite instability-high or mismatch repair deficiency (dMMR). Subsequently, the approval of NTRK inhibitors, designed to combat NTRK gene fusions prevalent in various tumor types, including pediatric cancers and adult solid tumors, further underscored the potential of tumor-agnostic therapies. The US Food and Drug Administration approvals of targeted therapies (BRAF V600E, RET fusion), immunotherapies (tumor mutational burden ≥10 mutations per megabase, dMMR) and an antibody-drug conjugate (Her2-positive–immunohistochemistry 3+ expression) with pan-cancer efficacy have continued, offering newfound hope to patients grappling with advanced solid tumors that harbor particular biomarkers. In this comprehensive review, the authors delve into the expansive landscape of tissue-agnostic targets and drugs, shedding light on the rationale underpinning this approach, the hurdles it faces, presently approved therapies, voices from the patient advocacy perspective, and the tantalizing prospects on the horizon. This is a welcome advance in oncology that transcends the boundaries of histology and location to provide personalized options.

肿瘤诊断疗法改变了根据肿瘤的起源或位置来确定肿瘤特征的传统方法,代表了肿瘤学的范式转变。取而代之的是,它们将目标锁定在助长恶性肿瘤生长的特定基因异常上。2017 年,美国食品药品管理局历史性地批准了免疫检查点抑制剂 pembrolizumab,这是肿瘤诊断疗法的分水岭。这一里程碑标志着基因组学与免疫学领域的联姻,一种基于基因组生物标志物,特别是微卫星不稳定性高或错配修复缺陷(dMMR)的免疫治疗药物获得了批准。随后,NTRK 抑制剂也获得了批准,这种抑制剂旨在对抗各种肿瘤类型(包括儿童癌症和成人实体瘤)中普遍存在的 NTRK 基因融合,进一步彰显了肿瘤诊断疗法的潜力。美国食品和药物管理局不断批准具有泛癌疗效的靶向疗法(BRAF V600E、RET融合)、免疫疗法(肿瘤突变负荷≥10个突变/兆碱基,dMMR)和抗体-药物共轭物(Her2阳性-免疫组化3+表达),为罹患携带特殊生物标记物的晚期实体瘤的患者带来了新的希望。在这篇综合性综述中,作者深入探讨了组织诊断靶点和药物的广阔前景,阐明了这种方法的基本原理、面临的障碍、目前已获批准的疗法、来自患者权益保护角度的声音以及地平线上诱人的前景。这是肿瘤学的一个可喜进步,它超越了组织学和位置的界限,提供了个性化的选择。
{"title":"The evolving landscape of tissue-agnostic therapies in precision oncology","authors":"Vivek Subbiah MD,&nbsp;Mohamed A. Gouda MD,&nbsp;Bettina Ryll MD, PhD,&nbsp;Howard A. Burris III MD,&nbsp;Razelle Kurzrock MD","doi":"10.3322/caac.21844","DOIUrl":"10.3322/caac.21844","url":null,"abstract":"<p>Tumor-agnostic therapies represent a paradigm shift in oncology by altering the traditional means of characterizing tumors based on their origin or location. Instead, they zero in on specific genetic anomalies responsible for fueling malignant growth. The watershed moment for tumor-agnostic therapies arrived in 2017, with the US Food and Drug Administration's historic approval of pembrolizumab, an immune checkpoint inhibitor. This milestone marked the <i>marriage</i> of genomics and immunology fields, as an immunotherapeutic agent gained approval based on genomic biomarkers, specifically, microsatellite instability-high or mismatch repair deficiency (dMMR). Subsequently, the approval of NTRK inhibitors, designed to combat <i>NTRK</i> gene fusions prevalent in various tumor types, including pediatric cancers and adult solid tumors, further underscored the potential of tumor-agnostic therapies. The US Food and Drug Administration approvals of targeted therapies (<i>BRAF</i> V600E, <i>RET</i> fusion), immunotherapies (tumor mutational burden ≥10 mutations per megabase, dMMR) and an antibody-drug conjugate (Her2-positive–immunohistochemistry 3+ expression) with pan-cancer efficacy have continued, offering newfound hope to patients grappling with advanced solid tumors that harbor particular biomarkers. In this comprehensive review, the authors delve into the expansive landscape of tissue-agnostic targets and drugs, shedding light on the rationale underpinning this approach, the hurdles it faces, presently approved therapies, voices from the patient advocacy perspective, and the tantalizing prospects on the horizon. This is a welcome advance in oncology that transcends the boundaries of histology and location to provide personalized options.</p>","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":null,"pages":null},"PeriodicalIF":503.1,"publicationDate":"2024-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.3322/caac.21844","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141174107","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
期刊
CA: A Cancer Journal for Clinicians
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