Christopher Tyson, Kevin H Li, Xiting Cao, James M O'Brien, Elliot K Fishman, Elizabeth K O'Donnell, Carlos Duran, Vijay Parthasarathy, Seema P Rego, Omair A Choudhry, Tomasz M Beer
{"title":"Tumor localization strategies of multicancer early detection tests: a quantitative assessment.","authors":"Christopher Tyson, Kevin H Li, Xiting Cao, James M O'Brien, Elliot K Fishman, Elizabeth K O'Donnell, Carlos Duran, Vijay Parthasarathy, Seema P Rego, Omair A Choudhry, Tomasz M Beer","doi":"10.1093/jncics/pkaf011","DOIUrl":"10.1093/jncics/pkaf011","url":null,"abstract":"<p><strong>Background: </strong>Multicancer early detection tests may expand cancer screening. Characterizing diagnostic resolution approaches following positive multicancer early detection tests is critical. Two trials employed distinct resolution approaches: a molecular signal to predict tissue of origin and an imaging-based diagnostic strategy. This modeling study characterizes diagnostic journeys and impact in a hypothetical population of average-risk multicancer early detection-eligible patients.</p><p><strong>Methods: </strong>A mathematical expression for diagnostic burden was derived using positive predictive value (PPV), molecular tissue of origin localization accuracy, and numbers of procedures associated with each diagnostic outcome. Imaging-based and molecular tissue of origin-informed strategies were compared. Excess lifetime cancer risk due to futile radiation exposure was estimated using organ-specific diagnostic imaging radiation doses.</p><p><strong>Results: </strong>Across all PPVs and localization performances, a molecular tissue of origin strategy resulted in a higher diagnostic burden (mean = 3.6 [0.445] procedures vs mean = 2.6 [0.100] procedures) for the imaging strategy. Estimated diagnostic burden was higher for molecular tissue of origin in 95.5% of all PPV and tissue of origin accuracy combinations; at least 79% PPV and 90% accuracy would be required for a molecular tissue of origin-informed strategy to be less burdensome than imaging. The maximum rate of excess cancer incidence from radiation exposure for multicancer early detection false-positive results (individuals aged 50-84 years) was 64.6 of 100 000 (annual testing, 99% specificity), 48.5 of 100 000 (biennial testing, 98.5% specificity), and 64.6 of 100 000 (biennial testing, 98% specificity).</p><p><strong>Conclusions: </strong>An imaging-based diagnostic strategy is more efficient than a molecular tissue of origin-informed approach across almost all PPV and tissue of origin accuracy combinations. The use of an imaging-based approach for cancer localization can be efficient and low-risk compared with a molecular-informed approach.</p>","PeriodicalId":14681,"journal":{"name":"JNCI Cancer Spectrum","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11897890/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143032964","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molly Scannell Bryan, Xiaohan Hu, Monika A Izano, Hina Mohammed, Marianna Wicks, Thomas Brown, George Simon, Henry Kaplan, Anna Berry
{"title":"Social determinants of health and variability in treatment for patients with early-stage non-small cell lung cancer.","authors":"Molly Scannell Bryan, Xiaohan Hu, Monika A Izano, Hina Mohammed, Marianna Wicks, Thomas Brown, George Simon, Henry Kaplan, Anna Berry","doi":"10.1093/jncics/pkae117","DOIUrl":"10.1093/jncics/pkae117","url":null,"abstract":"<p><strong>Background: </strong>In non-small cell lung cancer, social determinants of health (SDOH) influence treatment, but SDOH with geographic precision are infrequently used in real-world research because of privacy considerations. This research aims to characterize the influence of census tract-level SDOH on treatment for stage I and IIa non-small cell lung cancer.</p><p><strong>Methods: </strong>Patients diagnosed between January 1, 2017, and September 30, 2022, with stage I or IIa non-small cell lung cancer in the Syapse Learning Health Network had their addresses geocoded and linked to 6 census tract-level indicators of SDOH (the Centers for Disease Control and Prevention and Agency for Toxic Substances and Disease Registry Social Vulnerability Index, percentage housing burden, percentage broadband internet access, primary care shortage area, and rurality). Clinical and demographic characteristics were ascertained from medical records. Nested multinomial logistic regression models estimated associations between SDOH and initial treatment using 2-sided Wald tests. The collective statistical significance of SDOH was assessed using a likelihood ratio test comparing nested models. Descriptive statistics described time to treatment initiation.</p><p><strong>Results: </strong>Among 3595 patients, 58% were initially treated with surgery, 29% with radiation, and 12% with \"other.\" Two SDOH variables were associated with increased relative risk for radiation therapy compared with surgery: living in primary care shortage areas (relative risk = 1.61, 95% CI = 1.23 to 2.10) and living in nonmetropolitan areas (relative risk = 1.45, 95% CI = 1.02 to 2.07). The likelihood ratio test suggested that the 5 SDOH variables collectively improved the treatment model. Further, patients in areas with high Social Vulnerability Index, low internet access, and high housing burden initiated treatment later.</p><p><strong>Conclusion: </strong>When using precise estimates of geospatial SDOH, these measures were associated with treatment and should be considered in analyses of cancer outcomes.</p>","PeriodicalId":14681,"journal":{"name":"JNCI Cancer Spectrum","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11901590/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142709731","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Corinna Keeler, Nickilou Y Krigbaum, Barbara Cohn, Piera Cirillo
{"title":"Parental loss at age birth to 21 years and daughters' breast cancer and tumor characteristics.","authors":"Corinna Keeler, Nickilou Y Krigbaum, Barbara Cohn, Piera Cirillo","doi":"10.1093/jncics/pkaf004","DOIUrl":"10.1093/jncics/pkaf004","url":null,"abstract":"<p><strong>Background: </strong>Adverse events in childhood are linked to cancer risk across the life course, but evidence is lacking regarding parental death during childhood and breast cancer characteristics. We investigated whether women who experienced parental loss in childhood have a higher incidence of breast cancer and are at higher risk of aggressive disease.</p><p><strong>Methods: </strong>The Child Health and Development Studies (CHDS) consists of more than 15 000 families that enrolled during mothers' pregnancies between 1959 and 1967; family members were followed for cancer incidence and cause-specific mortality. We constructed an analytical cohort of all live-born CHDS daughters (N = 9169) linked to their parents' cause and date of death. We estimated adjusted hazard ratios of incident breast cancer, disease stage at diagnosis, and tumor hormone receptor expression for parental loss in Cox models adjusted for race, maternal breast cancer, and paternal age. Generalized linear models estimated associations between breast density and parental loss among a subsample of CHDS daughters (n = 610) with available mammography reports.</p><p><strong>Results: </strong>In total, 137 CHDS daughters were diagnosed with breast cancer by age 52 years, and 654 daughters had lost 1 or both parents when they were 21 years of age or younger. Loss of both parents was associated with breast cancer incidence (adjusted hazard ratio = 4.69, 95% CI = 1.68 to 13.04); late-stage disease at diagnosis (adjusted hazard ratio = 9.47, 95% CI = 1.38 to 64.84); and ERBB2 (formerly HER2)-positive, progesterone receptor-negative, and estrogen receptor-negative tumors. Loss of mother or father was associated with ERBB2-positive tumors. Breast density in the premenopause window was associated with loss of a mother or both parents.</p><p><strong>Conclusion: </strong>In a multigenerational cohort with well-defined cancer outcomes and validated cause-of-death data, life-course risk of breast cancer was 4.69 times higher among participants who had lost both parents during childhood. Subanalyses showed that parental loss was associated with late stage at diagnosis and tumor hormone markers of aggressive disease. Parental death during childhood could be added to medical histories to indicate a need for counseling on prevention and early detection of breast cancer.</p>","PeriodicalId":14681,"journal":{"name":"JNCI Cancer Spectrum","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11892429/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143005554","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Susanna C Larsson, Jie Chen, Xixian Ruan, Xue Li, Shuai Yuan
{"title":"Genome-wide association study and Mendelian randomization analyses reveal insights into bladder cancer etiology.","authors":"Susanna C Larsson, Jie Chen, Xixian Ruan, Xue Li, Shuai Yuan","doi":"10.1093/jncics/pkaf014","DOIUrl":"10.1093/jncics/pkaf014","url":null,"abstract":"<p><strong>Background: </strong>The causes of bladder cancer are not completely understood. Our objective was to identify blood proteins and modifiable causal risk factors for bladder cancer by combining genome-wide association study (GWAS) and Mendelian randomization (MR) analyses.</p><p><strong>Methods: </strong>We first performed a GWAS meta-analysis of 6984 bladder cancer case patients and 708 432 control individuals from 3 European databases. Next, we conducted 2-sample MR and colocalization analyses using data from the present GWAS and published GWAS meta-analyses on plasma proteins and modifiable factors.</p><p><strong>Results: </strong>Genome-wide association study meta-analysis uncovered 17 bladder cancer susceptibility loci, of which 3 loci were novel. Genes were enriched in pathways related to the metabolic and catabolic processes of xenobiotics and cellular detoxification. Proteome-wide MR analysis based on cis-acting genetic variants revealed that higher plasma levels of glutathione S-transferases were strongly associated with a reduced risk of bladder cancer. There is strong evidence of colocalization between GSTM1 and bladder cancer. Finally, multivariable MR analyses of suspected risk factors for bladder cancer revealed independent causal associations between smoking and adiposity, particularly abdominal obesity, and risk of bladder cancer.</p><p><strong>Conclusions: </strong>Findings from this large-scale GWAS and multivariable MR analyses highlight the key role of detoxification processes, particularly glutathione S-transferase 1, as well as smoking and abdominal obesity in bladder cancer etiology.</p>","PeriodicalId":14681,"journal":{"name":"JNCI Cancer Spectrum","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11950924/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143080080","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sarah Axeen, Alice J Chen, Darius N Lakdawalla, Neal Masia, Alexander Niyazov, Bhakti Arondekar, Stephen J Freedland
{"title":"Do trial benefits predict real-world gains in metastatic castration resistant prostate cancer.","authors":"Sarah Axeen, Alice J Chen, Darius N Lakdawalla, Neal Masia, Alexander Niyazov, Bhakti Arondekar, Stephen J Freedland","doi":"10.1093/jncics/pkaf018","DOIUrl":"10.1093/jncics/pkaf018","url":null,"abstract":"<p><strong>Background: </strong>It is important to understand the relationship between drug efficacy measured in randomized clinical trials (RCTs) and real-world drug effectiveness. We estimate how RCT overall survival (OS) and RCT radiographic progression-free survival (rPFS) benefits predict the association between treatments and real-world OS gains for metastatic castration-resistant prostate cancer (mCRPC) drugs.</p><p><strong>Methods: </strong>Using the National Cancer Institute list of approved cancer drugs and the National Comprehensive Cancer Network Treatment Guidelines, we identified all pharmaceutical therapies for mCRPC approved between 2010 and 2019. We obtained RCT OS and rPFS hazard ratios from the pivotal trials used for Food and Drug Administration (FDA) approval, and we estimated real-world OS hazard ratios using the Optum Clinformatics Extended DataMart Databases. We modeled real-world OS hazard ratios as a function of both RCT OS and RCT rPFS hazard ratios using Cox proportional hazards regressions, adjusted for year of diagnosis, age, race, and Elixhauser Comorbidity Index.</p><p><strong>Results: </strong>When we did not account for nonrandom real-world selection of patients into receiving a newly approved therapy (ie, \"treatment selection bias\"), real-world OS gains were 15% lower than associated RCT OS and RCT rPFS benefits. However, after accounting for treatment selection bias in real-world settings, real-world OS gains were almost 28% greater than RCT OS and RCT rPFS benefits. Association between treatment and OS gains increased the longer a new therapy was on the market.</p><p><strong>Conclusions: </strong>After adjusting for treatment selection bias, RCT OS and RCT rPFS estimates serve as useful, or even conservative, predictors of RW OS gains.</p>","PeriodicalId":14681,"journal":{"name":"JNCI Cancer Spectrum","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11879356/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143407954","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Isabel Arana, Raymond Liu, Lawrence Kushi, Erin Hahn, Meera Ragavan
{"title":"Screening for comprehensive social needs in patients with cancer: a narrative review.","authors":"Isabel Arana, Raymond Liu, Lawrence Kushi, Erin Hahn, Meera Ragavan","doi":"10.1093/jncics/pkaf012","DOIUrl":"10.1093/jncics/pkaf012","url":null,"abstract":"<p><strong>Background: </strong>Patients with cancer who report social needs have worse quality of life, lower health-care access, and suboptimal health outcomes. However, screening for social needs does not happen systematically, and successful screening tools, strategies, and workflows have seldom been described. The downstream effects of screening including resource navigation have also not been well characterized. The objective of this narrative review was to fill these gaps.</p><p><strong>Methods: </strong>Two investigators searched PubMed and Embase for studies that implemented a patient-facing social screening tool among patients with cancer between 2008 and 2023 using search terms including social screening, social needs, and cancer.</p><p><strong>Results: </strong>We identified 19 articles that met study inclusion criteria. The most common tool used was the validated Health Leads Social Toolkit. Most often, screening tools were administered electronically, sent directly to patients, and captured needs at a single time point during a patient's diagnosis. Screening response rates ranged between 10% and 60%. Less than half of the studies described downstream resource navigation for patients who screened positive for social needs. Only 1 study evaluated the impact of screening on clinical outcomes and quality of life. Screening for patients who do not speak English or who belong to historically racial, ethnic, and gender minority groups was limited.</p><p><strong>Conclusions: </strong>Screening for social needs has been shown to be feasible across delivery systems with numerous validated tools available. However, gaps remain in generalizability to diverse patient populations. Future work must identify how screening workflows can be successfully incorporated into routine clinical workflows.</p>","PeriodicalId":14681,"journal":{"name":"JNCI Cancer Spectrum","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11917213/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143052429","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jessica Y Islam, Yi Guo, Kea Turner, Amir Alishahi Tabriz, Yu Chen Lin, Denise C Vidot, Susan T Vadaparampil, Anna E Coghill, Marlene Camacho-Rivera, Gita Suneja
{"title":"Inequities in palliative care delivery to patients with HIV and stage IV cancers in the United States (2004-2020).","authors":"Jessica Y Islam, Yi Guo, Kea Turner, Amir Alishahi Tabriz, Yu Chen Lin, Denise C Vidot, Susan T Vadaparampil, Anna E Coghill, Marlene Camacho-Rivera, Gita Suneja","doi":"10.1093/jncics/pkae118","DOIUrl":"10.1093/jncics/pkae118","url":null,"abstract":"<p><strong>Background: </strong>People with HIV diagnosed with stage IV cancer are less likely to receive palliative care compared with those without HIV. Our objective was to evaluate inequities in palliative care receipt among people with HIV with stage IV cancer in the United States.</p><p><strong>Methods: </strong>We used the National Cancer Database (2004-2020), including adults (aged 18-89 years) with HIV with the 14 most common cancers that occur among people with HIV. Palliative care was defined as treatment provided with noncurative intent. Our main exposures included percent quartiles (Q) of adults without a high school degree (educational attainment) and median income quartiles within the patient's zip code. We used hierarchical multivariable Poisson regression to estimate adjusted prevalence ratios with 95% confidence intervals (CIs), adjusting for age, sex, year of diagnosis, race and ethnicity, and cancer type.</p><p><strong>Results: </strong>Among the included 10 120 people with HIV with stage IV cancer, 72% were men, 51% were either non-Hispanic Black or Hispanic or Latinx, 38% were aged 60 years and older, and 97% resided in urban areas; 14% received palliative care. Non-Hispanic Black people with HIV living in zip codes with lower quartiles of educational attainment were more likely to receive palliative care compared with those in the highest quartile (Q1 vs Q4: adjusted prevalence ratio = 1.93, 95% CI = 1.29 to 2.86). For income overall, compared with those in the highest quartile (Q4) of income, those in the lowest quartile had 26% higher likelihood of receiving palliative care (Q1 vs Q4: adjusted prevalence ratio = 1.26, 95% CI = 1.05 to 1.52), particularly among non-Hispanic Black adults (Q1 vs Q4: adjusted prevalence ratio = 1.67, 95% CI =1.25 to 2.22; Q2 vs Q4: adjusted prevalence ratio = 1.48, 95% CI = 1.09 to 2.01).</p><p><strong>Conclusions: </strong>Palliative care use among people with HIV with stage IV cancer is low. Contextual poverty plays a role in palliative care delivery to people with HIV and cancer, particularly among non-Hispanic Black people with HIV.</p>","PeriodicalId":14681,"journal":{"name":"JNCI Cancer Spectrum","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11897894/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142695536","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Muhammad Zaki Hidayatullah Fadlullah, David Nix, Cameron Herberts, Corinne Maurice-Dror, Alexander W Wyatt, Bogdana Schmidt, Brayden Fairbourn, Aik-Choon Tan, Liang Wang, Manish Kohli
{"title":"Multi-gene risk score for prediction of clinical outcomes in treatment-naïve metastatic castrate-resistant prostate cancer.","authors":"Muhammad Zaki Hidayatullah Fadlullah, David Nix, Cameron Herberts, Corinne Maurice-Dror, Alexander W Wyatt, Bogdana Schmidt, Brayden Fairbourn, Aik-Choon Tan, Liang Wang, Manish Kohli","doi":"10.1093/jncics/pkaf025","DOIUrl":"10.1093/jncics/pkaf025","url":null,"abstract":"<p><strong>Background: </strong>To determine the performance of a multi-gene copy number variation (MG-CNV) risk score in metastatic tissue and plasma biospecimens from treatment-naïve metastatic castration-resistant prostate cancer (mCRPC) patients for prediction of clinical outcomes.</p><p><strong>Methods: </strong>The mCRPC tissue and plasma cell-free DNA (cfDNA) biospecimen sequencing results obtained from publicly accessed cohorts in dbGaP, cBioPortal, and an institutional mCRPC cohort were used to develop a MG-CNV risk score derived from gains in AR, MYC, COL22A1, PIK3CA, PIK3CB, NOTCH1 and losses in TMPRSS2, NCOR1, ZBTB16, TP53, NKX3-1 in independent cohorts for determining overall survival (OS), progression-free survival (PFS) to first-line androgen receptor pathway inhibitors (ARPIs). The range of the risk scores for each cohort was dichotomized into \"high-risk\" and \"low-risk\" groups and association with OS/PFS determined. Univariate and multivariable Cox proportional hazards regressions were applied for survival analyses (P < .05 for statistical significance).</p><p><strong>Results: </strong>Of 1137 metastatic tissue-plasma biospecimens across all cohorts, 699/1137 were treatment-naive mCRPC (235/699 metastatic tissue; 464/699 plasma-cfDNA), and 311/1137 were matched tissue-cfDNA pairs. In multivariable analysis, the MG-CNV risk score derived from metastatic tissue or in cfDNA was statistically significantly associated with OS with high score associated with short survival (hazard ratio = 2.65, confidence interval = 1.99 to 3.51; P = 1.35-11) and shorter PFS to ARPIs (median PFS of 7.8 months) compared with 14 months in patients with low-risk score.</p><p><strong>Conclusions: </strong>A molecular risk score in treatment-naïve mCRPC state obtained either in metastatic tissue or cfDNA predicts clinical survival outcomes and offers a tumor biology-based tool to design biomarker-based enrichment clinical trials.</p>","PeriodicalId":14681,"journal":{"name":"JNCI Cancer Spectrum","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11954629/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143556887","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Toru Yoshino, Zao Zhang, Ryota Sato, Stanley Lipkowitz, Takeo Fujii
{"title":"Revisiting surrogacy of pathological complete response for long-term survival in triple-negative breast cancer.","authors":"Toru Yoshino, Zao Zhang, Ryota Sato, Stanley Lipkowitz, Takeo Fujii","doi":"10.1093/jncics/pkaf022","DOIUrl":"10.1093/jncics/pkaf022","url":null,"abstract":"<p><strong>Background: </strong>Pathological complete response has been used as a primary endpoint in neoadjuvant trials in early stage triple-negative breast cancer, and the Food and Drug Administration accepted pathological complete response as a surrogate endpoint for long-term survival outcomes in high-risk early stage breast cancer for new drug approval. However, there is insufficient trial-level data to robustly support pathological complete response as a surrogate for long-term survival in triple-negative breast cancer.</p><p><strong>Methods: </strong>A systematic literature review was performed to identify randomized clinical trials of neoadjuvant systemic therapy for patients with clinical stage I-III triple-negative breast cancer. Data of odds ratios (ORs) for pathological complete response and hazard ratios (HRs) for event-free survival and overall survival were extracted. Disease-free survival was used as an alternative when event-free survival data were unavailable. A linear regression model on a logarithmic scale, coefficient of difference, and 95% confidential interval (CI) were calculated to assess the trial-level association between odds ratio for pathological complete response and hazard ratio for overall survival and event-free survival.</p><p><strong>Results: </strong>Eight trials with 2342 patients were included. Three trials tested immune checkpoint inhibitors. Coefficient of difference (R2) was 0.2 for hazard ratio of event-free survival (95% CI = 0.17 to 0.22, P = .27), and R2 for hazard ratio of overall survival was 0.19 (95% CI = 0.17 to 0.22, P = .33).</p><p><strong>Conclusions: </strong>There is no strong evidence to support using pathological complete response as a surrogate marker for event-free survival or overall survival in early stage triple-negative breast cancer at the trial level. Because of the necessity of minimizing drug approval delay with reliable long-term outcome, further studies of surrogate markers in early stage triple-negative breast cancer are warranted.</p>","PeriodicalId":14681,"journal":{"name":"JNCI Cancer Spectrum","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11932712/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143483006","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ozlem Aynaci, Yetkin Tuac, Layth Mula-Hussain, Lubna Hammoudeh, Salameh Obeidat, Enas Abu Abeelh, Ahmed H Ibrahim, Sepideh Mohammadipour, Bader Alali, Ahmed Jdaini, Ali Barki, Nesrine Mejri, Zeinab Alhaddad, Nadeem Pervez, Hussain Al Hussain, Mohamad Kadri, Mohamed A Elfagieh, Adda Bounedjar, Moamin Junaid, Ahmed M Badheeb, Ibrahim Abu Ghida, Shalini Moningi, Jonathan E Leeman, Peter F Orio, Paul L Nguyen, Anthony V D'Amico, Mutlay Sayan
{"title":"Prostate cancer screening in the Middle East and North Africa: a cross-sectional study on current practices.","authors":"Ozlem Aynaci, Yetkin Tuac, Layth Mula-Hussain, Lubna Hammoudeh, Salameh Obeidat, Enas Abu Abeelh, Ahmed H Ibrahim, Sepideh Mohammadipour, Bader Alali, Ahmed Jdaini, Ali Barki, Nesrine Mejri, Zeinab Alhaddad, Nadeem Pervez, Hussain Al Hussain, Mohamad Kadri, Mohamed A Elfagieh, Adda Bounedjar, Moamin Junaid, Ahmed M Badheeb, Ibrahim Abu Ghida, Shalini Moningi, Jonathan E Leeman, Peter F Orio, Paul L Nguyen, Anthony V D'Amico, Mutlay Sayan","doi":"10.1093/jncics/pkaf019","DOIUrl":"10.1093/jncics/pkaf019","url":null,"abstract":"<p><strong>Background: </strong>Prostate cancer is a substantial health concern in the Middle East and North Africa region, with many cases diagnosed at advanced stages, a high mortality to incidence ratio, and low prostate cancer awareness. This study aimed to evaluate prostate cancer screening practices in the region to inform effective early detection and management strategies.</p><p><strong>Methods: </strong>A cross-sectional survey was conducted from July 1, 2023, to November 8, 2024, among physicians from 19 countries in the Middle East and North Africa region. The study used a validated questionnaire to assess prostate cancer screening practices, barriers, and educational needs.</p><p><strong>Results: </strong>The survey had a response rate of 96.8% and 1163 participants. Of these participants, 34.7% routinely performed prostate cancer screenings, with 61.1% using prostate-specific antigen tests. The primary barrier was lack of patient awareness (51.2%). In addition, 65.3% of participants had no formal training. To improve screening rates, participants suggested better patient education (63.5%), increased training for health-care professionals (41.9%), and improved access to screening equipment (38.9%).</p><p><strong>Conclusion: </strong>This study revealed that prostate cancer screening rates were low, with barriers including a lack of patient awareness and formal training among physicians. Addressing these issues through culturally tailored education programs may improve early detection rates and ultimately reduce the burden of prostate cancer in the Middle East and North Africa region.</p>","PeriodicalId":14681,"journal":{"name":"JNCI Cancer Spectrum","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11927531/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143373974","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}