{"title":"Learning from prostate cancer statistics","authors":"Ruth Etzioni, Lukas Owens","doi":"10.3322/caac.70037","DOIUrl":"https://doi.org/10.3322/caac.70037","url":null,"abstract":"<p>The population represents the ultimate uncontrolled experiment, yet data on cancer statistics provide an opportunity to learn about real-world outcomes of cancer control activities and policies. In the case of prostate cancer, population data have been critically important in generating and confirming hypotheses about the impacts of screening and treatment advances on the population burden of the disease. Tracking prostate cancer statistics—incidence, mortality, and survival—and how they change over time is thus a prerequisite for understanding the success (or lack thereof) of efforts to control this most common cancer in American men. But population statistics are multifactorial; explaining them requires also considering their many potential drivers and the mechanisms by which disease control efforts play out in the population.</p>\u0000<p>Consider the example of prostate cancer incidence, prominently reported in this issue’s update on prostate cancer statistics.<span><sup>1</sup></span> Prostate cancer incidence is influenced by prostate-specific antigen (PSA) screening rates in the population. Incidence increased dramatically during the early years of the PSA screening era, prompting concerns that screening was leading to overdiagnosis. Although overdiagnosis did indeed turn out to be a problematic outcome of screening, work by Feuer and Wun<span><sup>2</sup></span> in the early 1990s assured that increases in disease incidence were to be expected when a new screening test was adopted at the population level. The mechanism—initial depletion of the prevalent pool of cases by the screening test—leads to a predicted peak in incidence followed by declines because of the absence in the prevalent pool of those previously detected cases. Feuer and Wun demonstrated that the height and duration of the peak would be driven by the lead time, which is the time by which screening advances disease diagnosis. The lead time is critical not only in the timing of incidence swings after the adoption of screening but also in the delay until any effects of screening on disease mortality are observed. And the average lead time associated with prostate cancer screening is not short—estimates based on the first decade of PSA screening place the mean lead time between 5 and 7 years.<span><sup>3</sup></span></p>\u0000<p>The update of prostate cancer statistics in this issue of <i>CA: A Cancer Journal for Clinicians</i> highlights more recent incidence trends, specifically the persistence of recent increases overall and in advanced-stage disease. These trends have generated concern because they are what one would expect in a population abandoning screening. Indeed, studies tracking both incidence and screening patterns have been on the alert for such trends, particularly after the issuance of the D recommendation against routine prostate cancer screening for all ages by the US Preventive Services Task Force in 2012.<span><sup>4</sup></span> Although some modest reductions in p","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":"4 1","pages":""},"PeriodicalIF":254.7,"publicationDate":"2025-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145183267","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}
Hetal D. Mistry, MacKenzie R. Adams, Charu Taneja, Lauren J. Massingham, Elizabeth H. Dibble, Kara L. Leonard, Jesse Hart, Galina G. Lagos, Mary Anne Fenton
{"title":"A patient with newly diagnosed breast cancer found to have mosaic TP53 likely pathogenic variant","authors":"Hetal D. Mistry, MacKenzie R. Adams, Charu Taneja, Lauren J. Massingham, Elizabeth H. Dibble, Kara L. Leonard, Jesse Hart, Galina G. Lagos, Mary Anne Fenton","doi":"10.3322/caac.70034","DOIUrl":"https://doi.org/10.3322/caac.70034","url":null,"abstract":"<h2> CASE OVERVIEW</h2>\u0000<p>A 36-year-old, nulligravid woman with a history of controlled eosinophilic esophagitis, asthma, and dense fibrocystic breasts was referred to the Breast Health Center after abnormal screening mammography. She reported recent fatigue and intermittent diarrhea but had a negative colonoscopy and food allergy testing this year.</p>\u0000<p>Her family history was significant for a maternal grandmother with a question of uterine cancer, a maternal grandfather who died of renal cancer at age 62 years, a paternal grandmother who died of brain cancer at age 56 years, a paternal grandfather who died of brain cancer at age 80 years, and a history of premalignant changes in the esophagus in her father. She has never smoked tobacco and consumes zero to three drinks of alcohol weekly. She has been physically active in multiple sports, including biking and swimming, and played the cello.</p>\u0000<p>Mammography demonstrated extremely dense breast tissue with new calcifications in both breasts. Diagnostic mammography demonstrated indeterminate grouped calcifications spanning 19 mm in the right breast at 5 o'clock and 7 mm in the left breast 12 o'clock. She subsequently underwent stereotactic needle biopsies of both areas, which revealed right breast ductal carcinoma in situ (DCIS), nuclear grade 3, with 90% estrogen receptor (ER) expression; and left ductal carcinoma, nuclear grade 3, with 95% ER expression, 40% progesterone receptor (PR) expression, negative human epidermal growth factor receptor 2 (HER-2) status, and a Ki-67 index of 40%. Within the left breast biopsy there was an absence of myoepithelial cells, raising concern that the findings reflected an unusual type of invasive carcinoma. She then underwent bilateral breast magnetic resonance imaging (MRI), which revealed the known areas of DCIS in the bilateral breasts and also revealed a 1.0-cm mass inferior to the left breast DCIS at 12 o'clock that was considered suspicious. On subsequent ultrasound, it corresponded to an 8-mm mass in the left breast at 10 o'clock. A biopsy of the mass demonstrated a spindle cell tumor, favoring malignant phyllodes tumor with pleomorphic liposarcomatous differentiation that was negative for ER, PR, and HER2 (triple-negative), with a Ki-67 index of 30%.</p>\u0000<p>At her multidisciplinary consultation, she was referred for genetic evaluation having met National Comprehensive Cancer Network (NCCN) criteria based on her diagnosis of breast cancer when younger than 50 years.<span><sup>1</sup></span> Given the concern for invasive left breast cancer and possible need for chemotherapy, she was also referred for fertility preservation.</p>\u0000<p>She completed a comprehensive 76-gene germline genetic test, which revealed tumor protein p53 (<i>TP53)</i> likely pathogenic variant (LPV; c.716A>G; p.N239S). Pathogenic variants (PVs) and LPVs are DNA sequence changes that are associated with increased risk of disease. There is well established evidence that PVs are di","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":"13 1","pages":""},"PeriodicalIF":254.7,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145127733","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}
{"title":"Advances in pancreatic cancer early diagnosis, prevention, and treatment: The past, the present, and the future","authors":"Alessandro Mannucci, Ajay Goel","doi":"10.3322/caac.70035","DOIUrl":"https://doi.org/10.3322/caac.70035","url":null,"abstract":"Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a dismal prognosis, largely because of late-stage diagnosis and therapeutic resistance. PDAC incidence has been rising, with modifiable and non-modifiable risk factors contributing to disease development. Chronic pancreatitis, diabetes mellitus, smoking, obesity, and familial predisposition have been implicated in PDAC pathogenesis. Early clinical manifestations are vague and insidious; therefore, PDAC is often diagnosed at an advanced stage, limiting curative treatment options. Efforts to improve early detection have focused on serum biomarkers (e.g., carbohydrate antigen 19-9), imaging modalities, and liquid biopsies. Endoscopic ultrasound and magnetic resonance imaging have demonstrated potential in identifying early-stage disease in certain high-risk populations. Surgical resection remains the only potentially curative option, but only 15%–20% of patients have resectable disease at diagnosis. Neoadjuvant chemotherapy has emerged as a promising strategy to improve resectability and survival outcomes. For patients with locally advanced or metastatic PDAC, combination chemotherapy regimens such as FOLFIRINOX (folinic acid, 5-fluorouracil, irinotecan, and oxaliplatin), NALIRIFOX (5-fluorouracil, oxaliplatin, liposomal irinotecan, and leucovorin), and combined gemcitabine/nanoparticle albumen-bound paclitaxel offer survival benefits, although toxicity remains a concern, especially for platinum-based therapies. Several breakthroughs in molecular profiling have led to the development of targeted therapies, including sotorasib and olaparib. Immunotherapy has shown limited success in PDAC due to its immunosuppressive tumor microenvironment. However, novel combination approaches are under investigation, including quadruplet therapy, immune checkpoint inhibitors with oncolytic viruses, stromal-targeting agents, and personalized neoantigen vaccines. Key priorities for future research include identifying reliable biomarkers for early detection, refining patient selection for targeted therapies, and developing innovative strategies to overcome treatment resistance.","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":"72 1","pages":""},"PeriodicalIF":254.7,"publicationDate":"2025-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145083925","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}
{"title":"Reviewer acknowledgement 2025","authors":"","doi":"10.3322/caac.70033","DOIUrl":"https://doi.org/10.3322/caac.70033","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, 2024, to June 30, 2025. These individuals go beyond expectations by consistently and expeditiously delivering comprehensive, discerning reviews.</p>\u0000<p>Lauren Antrim</p>\u0000<p>Pedro Barata</p>\u0000<p>Nabila Bennani</p>\u0000<p>Ari Birnbaum</p>\u0000<p>Sue Bornstein</p>\u0000<p>Donald Cannon</p>\u0000<p>Michael Carney</p>\u0000<p>Elizabeth Cathcart-Rake</p>\u0000<p>Annie Chan</p>\u0000<p>Huizi Chen</p>\u0000<p>Carissa Chu</p>\u0000<p>Sean David</p>\u0000<p>Ruth Etzioni</p>\u0000<p>Robert Ferris</p>\u0000<p>Courtney Finlayson</p>\u0000<p>Bryan Fuchs</p>\u0000<p>Elizabeth Garrett-Mayer</p>\u0000<p>William Hall</p>\u0000<p>Michael Halpern</p>\u0000<p>Ole-Petter Hamnvik</p>\u0000<p>David Hui</p>\u0000<p>Linda Jacobs</p>\u0000<p>Salvador Jaime-Casas</p>\u0000<p>Rebecca Johnson</p>\u0000<p>Corinne Joshu</p>\u0000<p>David Keefe</p>\u0000<p>WonSeog Kim</p>\u0000<p>Elise Kohn</p>\u0000<p>Lindsay M. Kuroki</p>\u0000<p>Rita Kuwahara</p>\u0000<p>Richard Lee</p>\u0000<p>Shing Lee</p>\u0000<p>Phebe Lemert</p>\u0000<p>Stanley Liauw</p>\u0000<p>Stephen Liu</p>\u0000<p>Shail Maingi</p>\u0000<p>Sendurai Mani</p>\u0000<p>Jonathan Marron</p>\u0000<p>Brittany C. McGill</p>\u0000<p>Jacob Miller</p>\u0000<p>Paul Montgomery</p>\u0000<p>Susan O'Brien</p>\u0000<p>Krishnan Patel</p>\u0000<p>Rodolfo Alberto Rey</p>\u0000<p>Tina Rizack</p>\u0000<p>Nabil Saba</p>\u0000<p>Stephanie Smith</p>\u0000<p>Umang Swami</p>\u0000<p>Wade Swenson</p>\u0000<p>Russell Taichman</p>\u0000<p>Yungan Tao</p>\u0000<p>Molly Taylor</p>\u0000<p>Ayalew Tefferi</p>\u0000<p>Sarah Temkin</p>\u0000<p>William Tew</p>\u0000<p>Premal Thaker</p>\u0000<p>Jonathan Thompson</p>\u0000<p>Michael Thun</p>\u0000<p>Katherine Tossas</p>\u0000<p>Zaza Tsitsishvili</p>\u0000<p>Adam Wahida</p>\u0000<p>Thomas Zilli</p>\u0000<p>Miguel Zugman</p>","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":"121 1","pages":""},"PeriodicalIF":254.7,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145078376","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}
{"title":"Correction to \"Cancer treatment and survivorship statistics, 2025\".","authors":"","doi":"10.3322/caac.70036","DOIUrl":"https://doi.org/10.3322/caac.70036","url":null,"abstract":"","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":"3 1","pages":""},"PeriodicalIF":254.7,"publicationDate":"2025-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145043807","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}
{"title":"Fertility counseling for reproductive-age women with cancer should address gestational carriers","authors":"Carrie Printz","doi":"10.3322/caac.70031","DOIUrl":"10.3322/caac.70031","url":null,"abstract":"<p>Major cancer organizations, including the American Society of Clinical Oncology and the National Comprehensive Cancer Network, recommend that all reproductive-age women with cancer should receive fertility counseling. Certain cancer treatments, particularly those for women with breast or gynecological cancers, can affect fertility and make it more difficult for women to conceive. Upon receiving fertility counseling before cancer treatment, some patients may decide to freeze their oocytes or embryos for future in vitro fertilization (IVF). Others may not seek family planning help until after receiving chemotherapy or pelvic radiation, which can damage their ability to have biological children.</p><p>Women in both groups may turn to gestational carriers, also called surrogates, to carry a child for them through IVF. Gestational carriers may be the only option for patients whose uterus was removed or damaged during cancer treatment.</p><p>In a study published in <i>Cancer</i> (doi:10.1002/cncr.35844), researchers sought to further characterize the role of gestational carriers among reproductive-age women diagnosed with cancer. The study assessed which patients used gestational carriers and how often, as well as their pregnancy outcomes.</p><p>Researchers used data from eight statewide cancer registries linked with the Society for Assistive Reproductive Technology Clinic Outcomes Reporting System (SART CORS). Reporting IVF cycles is federally mandated, and SART CORS includes information from more than 90% of US clinics. Drawing on these data, investigators assessed a retrospective cohort of women with a cancer diagnosis who subsequently initiated IVF procedures from 2004 to 2018.</p><p>The statewide cancer registries were in Arizona, California, Colorado, Massachusetts, Maryland, North Carolina, New York, and Virginia. Each state has a high number of IVF cycles performed annually and 1000 or more IVF-assisted births each year.</p><p>Modified Poisson models were used to estimate prevalence rates and 95% confidence intervals (CI). Discrete Cox regression models were used to calculate the hazard ratio and CI. Multivariable models were adjusted for age, state, and calendar year.</p><p>Of the 1095 women diagnosed with cancer who used IVF with cryopreserved embryos or oocytes, 19.1% worked with a gestational carrier. Women involved gestational carriers more often when they were initiating IVF for fertility preservation rather than after their cancer treatment. Those who had chemotherapy versus no chemotherapy also were more likely to use gestational carriers. In addition, the use of donor oocytes or embryos was more common in women who worked with a gestational carrier.</p><p>In the sample, 156 women were diagnosed with gynecologic cancer, with 89 having possibly threatened fertility related to surgery or pelvic radiation. In this group, 14.6% (<i>n</i> = 13) initiated IVF before treatment, with 12 ultimately using a gestational carrier. In the group th","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":"75 5","pages":"365-367"},"PeriodicalIF":232.4,"publicationDate":"2025-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://acsjournals.onlinelibrary.wiley.com/doi/epdf/10.3322/caac.70031","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145038285","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}
{"title":"Regorafenib as maintenance therapy showed significant delayed progression of non-adipocytic soft tissue sarcomas","authors":"Carrie Printz","doi":"10.3322/caac.70032","DOIUrl":"10.3322/caac.70032","url":null,"abstract":"<p>Because advanced non-adipocytic soft tissue sarcomas (STSs) are aggressive with poor outcomes, the goal of treatment is to delay progression and ensure quality of life. Doxorubicin (an anthracycline chemotherapy) is first-line standard of care for these patients. Researchers have discussed whether adding maintenance therapy might improve progression-free survival (PFS), but few studies have examined the option.</p><p>Non-adipocytic STSs are cancers that start in the body’s connective tissues but not in fat cells (which are called adipocytes).</p><p>EREMISS, a double-blind, randomized, controlled, phase 2 trial, evaluated the safety and PFS of maintenance therapy with regorafenib in patients with non-adipocytic STSs. Findings showed that regorafenib significantly increased PFS after first-line treatment by a median difference of 2.1 months. Results were published in the <i>Annals of Oncology</i> (doi:10.1016/j.annonc.2025.03.024).</p><p>“The sarcoma community has had a lot of conversation around the role of maintenance, and I was really pleased to see this well-designed study,” says Lara Davis, MD, an associate professor of medicine in the Division of Hematology/Medical Oncology and the director of the Sarcoma Program at Oregon Health & Science University in Portland. “The findings are clearly significant and look very promising. While the role of maintenance therapy in general is not fully established, it’s absolutely worth additional investigation.”</p><p>The EREMISS trial assessed the activity and safety of regorafenib in 126 patients with advanced non-adipocytic STSs who were enrolled in 17 centers in France from May 2019 to November 2022.</p><p>Regorafenib is an oral multikinase inhibitor that can target oncogenic pathways. Studies have shown its effectiveness in treating some sarcoma subtypes, aside from liposarcomas.</p><p>Female patients represented 55% of the total enrollment, and the median age was 58 years. The most common subtype was leiomyosarcoma (59%).</p><p>Participants who had stable disease or a partial response after six cycles of doxorubicin as a first-line treatment received 120 mg of regorafenib per day for 3 weeks on and 1 week off. The primary end point was PFS. It was accessible in 122 patients.</p><p>The median PFS by blinded central review was 3.5 months in the placebo arm and 5.6 months in the regorafenib arm. Although overall survival was not the primary end point, it was 20.5 months for the placebo group and 27.6 months for the regorafenib group.</p><p>In the placebo arm, 4.8% of the patients experienced grade 3 or higher adverse events, whereas 56.3% of the patients receiving regorafenib did. The most common adverse events were asthenia (9%), atrial hypertension (8%), and rash (8%).</p><p>Robin Jones, MD, a medical oncologist and sarcoma specialist at the Royal Marsden Cancer Center in London, praises the study’s design.</p><p>“The authors should be congratulated for actually completing and publishing the tria","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":"75 5","pages":"368-370"},"PeriodicalIF":232.4,"publicationDate":"2025-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://acsjournals.onlinelibrary.wiley.com/doi/epdf/10.3322/caac.70032","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145038286","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}
Stephanie M Smith,Lauren C Heathcote,Jennifer N John,Jasan Zimmerman,Catherine Benedict,Abby R Rosenberg,Lidia Schapira
{"title":"Talking with adolescent and young adult cancer survivors about health after cancer: A review and communication guide for clinicians.","authors":"Stephanie M Smith,Lauren C Heathcote,Jennifer N John,Jasan Zimmerman,Catherine Benedict,Abby R Rosenberg,Lidia Schapira","doi":"10.3322/caac.70030","DOIUrl":"https://doi.org/10.3322/caac.70030","url":null,"abstract":"Adolescent and young adult (AYA) cancer survivors represent a vulnerable population in cancer care and survivorship. AYA survivors are a heterogeneous group that includes people between the ages of 15 and 39 years who were treated for cancer during their childhood or AYA years, at which time they had variable agency and may have received cancer care in pediatric or adult settings. AYA survivors experience one or multiple health care transitions, moving from active oncology to posttreatment survivorship and/or from pediatric to adult care. Clinician communication that centers the needs and preferences of the AYA and their family (parent, partner, other support person) is a therapeutic tool that can support AYAs in these health care transitions and promote AYA engagement in their care. In this article, the authors review clinician communication practices through the lens of AYAs' and families' lived experiences with a focus on the initial diagnosis and treatment phase, completion of treatment, and throughout posttreatment survivorship care. Specific communication topics relevant to survivorship encompass managing uncertainty and fear of cancer recurrence, discussing treatment-related future health risks, and supporting self-management and engagement in care. Best practices for clinician communication include maintaining openness, compassion, and flexibility to re-assess and adapt communication styles as an AYA cancer survivors' needs, concerns, and preferences change over time.","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":"19 1","pages":""},"PeriodicalIF":254.7,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144960291","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}
Tyler B Kratzer, Natalia Mazzitelli, Jessica Star, William L Dahut, Ahmedin Jemal, Rebecca L Siegel
{"title":"Prostate cancer statistics, 2025.","authors":"Tyler B Kratzer, Natalia Mazzitelli, Jessica Star, William L Dahut, Ahmedin Jemal, Rebecca L Siegel","doi":"10.3322/caac.70028","DOIUrl":"https://doi.org/10.3322/caac.70028","url":null,"abstract":"<p><p>Prostate cancer is the most common cancer among men in the United States, and the incidence of advanced disease is increasing rapidly. This article provides an overview of prostate cancer occurrence using population-based incidence and mortality data from the National Cancer Institute and the Centers for Disease Control and Prevention. Prostate cancer incidence trends have reversed from a decline of 6.4% per year during 2007 through 2014 to an increase of 3.0% annually during 2014 through 2021. The increasing trend is confined to distant-stage disease in men younger than 55 years and to regional/distant-stage disease in men aged 55-69 years but includes early stage disease in men aged 70 years and older. Over the past decade of data, distant-stage disease has increased by 2.6% annually in men younger than 55 years, 6.0% annually in men aged 55-69 years, and 6.2% annually in men aged 70 years and older. American Indian/Alaska Native, Asian American/Pacific Islander, and Hispanic men are less likely than Black and White men to be diagnosed with localized disease (64%-67% vs. 71%-72%). Compared with White men, American Indian/Alaska Native men have 12% higher prostate cancer mortality despite 13% lower incidence, whereas Black men have double the prostate cancer mortality, with 67% higher incidence. In summary, continued increases in the diagnosis of advanced prostate cancer and persistent racial disparities underscore the need for redoubled efforts to optimize early detection while limiting overdiagnosis and to understand and address barriers to equitable outcomes.</p>","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":" ","pages":""},"PeriodicalIF":232.4,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144937083","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}
Gwendolyn P Quinn,Michelle Bayefsky,Brooke Cherven,Lauren V Ghazal,Kara N Goldman,Nina Francis Levin,Leena Nahata,Bobby Najari,Sameer Thakker,Daniel R Greenberg,Joshua A Halpern,Susan T Vadaparampil
{"title":"Update and new advances in fertility preservation and cancer.","authors":"Gwendolyn P Quinn,Michelle Bayefsky,Brooke Cherven,Lauren V Ghazal,Kara N Goldman,Nina Francis Levin,Leena Nahata,Bobby Najari,Sameer Thakker,Daniel R Greenberg,Joshua A Halpern,Susan T Vadaparampil","doi":"10.3322/caac.70029","DOIUrl":"https://doi.org/10.3322/caac.70029","url":null,"abstract":"Adolescents and young adults with a cancer diagnosis face unique challenges during treatment and into survivorship related to fertility and family building. This review provides an updated overview of the impact of cancer and its associated treatments, including novel treatments in male and female fertility. An overview of fertility preservation and family building options, including experimental options, is also provided.","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":"19 1","pages":""},"PeriodicalIF":254.7,"publicationDate":"2025-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144825550","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}