1
|
Subbiah V, Othus M, Palma J, Cuglievan B, Kurzrock R. Designing Clinical Trials for Patients With Rare Cancers: Connecting the Zebras. Am Soc Clin Oncol Educ Book 2025; 45:e100051. [PMID: 40228175 DOI: 10.1200/edbk-25-100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
The field of rare cancer research is rapidly transforming, marked by significant progress in clinical trials and treatment strategies. Rare cancers, as defined by the National Cancer Institute, occur in fewer than 150 cases per million people each year, yet they collectively represent a significant portion of all cancer diagnoses. Because of their infrequency, these cancers pose distinct challenges for clinical trials, including limited patient populations, geographical dispersion, and a general lack of awareness of treatment options. Economic limitations further complicate drug development, making initiatives such as the Orphan Drug Act essential for incentivizing research. The advent of next-generation sequencing (NGS) and precision medicine has been instrumental in identifying actionable genetic alterations in parallel with an explosion in the development of genomically targeted therapies, immunotherapies, and antibody drug conjugates. Advances in clinical NGS, precision medicine, and tumor-agnostic therapies have become central to the progress in rare cancer research. The development and approval of tumor-agnostic drugs, such as BRAF, NTRK, and RET inhibitors, and immunotherapy for mismatch repair deficient/microsatellite instability-high status cancers highlight the potential of personalized treatments across diverse cancer types and across the age spectrum. Collaborative trials from cooperative groups including SWOG DART, ASCO TAPUR, NCI-MATCH, pediatric COG-match, DRUP, IMPRESS, and innovative registrational basket and platform trials (eg, VE-Basket, ROAR, LIBRETTO-001, ARROW), along with patient advocacy group-run trials like TRACK, are enhancing access to clinical trials. In addition, artificial intelligence has the potential to improve the trial matching process. An integrated approach, combining these innovations in collaboration with multiple stakeholders, is crucial for advancing rare cancer research, offering hope for better patient outcomes and quality of life.
Collapse
Affiliation(s)
| | - Megan Othus
- SWOG Cancer Research Network Statistical Center, Seattle, WA
- Division of Public Health, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jim Palma
- TargetCancer Foundation, Rare Cancer Patient Advocacy Group, Cambridge, MA
| | - Branko Cuglievan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Razelle Kurzrock
- Genomic Sciences and Precision Medicine Center, and Medical College of Wisconsin Cancer Center, Milwaukee, WI
- WIN Consortium, Paris, France
- University of Nebraska, Lincoln, NE
| |
Collapse
|
2
|
Sepulveda G. Waging peace: soothing the war metaphor about health and disease. Med Confl Surviv 2025; 41:18-25. [PMID: 39803826 DOI: 10.1080/13623699.2025.2450328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Affiliation(s)
- Gabriel Sepulveda
- Centro de Estudios en Neurociencia Humana y Neuropsicología, Programa de Doctorado en Psicología, Universidad Diego Portales, Santiago, Chile
| |
Collapse
|
3
|
Storandt MH, Shi Q, Eng C, Lieu C, George T, Stoppler MC, Mauer E, Yilma B, Fragkogianni S, Teslow EA, Mahipal A, Jin Z. Genomic Landscapes of Early-Onset Versus Average-Onset Colorectal Cancer Populations. Cancers (Basel) 2025; 17:836. [PMID: 40075683 PMCID: PMC11899610 DOI: 10.3390/cancers17050836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Rates of early-onset colorectal cancer (eoCRC), defined as disease diagnosed at <50 years of age, are increasing. The incidence and spectrum of somatic and pathogenic germline variants (PGV) in this population are not well understood. METHODS This cross-sectional study leveraged Tempus' clinicogenomic database, including de-identified records of patients diagnosed with CRC between 2000-2022, to analyze and compare eoCRC and average-onset colorectal cancer (aoCRC, disease diagnosed ≥50 years of age) patients. The frequency and spectrum of somatic mutations and PGVs in patients with eoCRC and aoCRC were evaluated and compared. RESULTS Among 11,006 participants in this study, 57% were male, 76% were white, and 80% had stage 4 disease. Within the total cohort, 2379 had eoCRC and 8627 had aoCRC. Among patients with eoCRC, 4.2% had a tumor with high microsatellite instability and/or deficient mismatch repair (MSI-H/dMMR) and 6.8% with aoCRC had an MSI-H/dMMR tumor (p < 0.001). The most frequent somatic mutations involved TP53, APC, and KRAS, with the most significant difference in BRAF, which was more frequently mutated in aoCRC (9.8% vs. 4.7%, p < 0.0001). In total, 1413 (59.4%) eoCRC and 4898 (56.8%) aoCRC patients had matched normal specimen (blood or saliva) sequencing and a PGV was identified in 6.9% of eoCRC and 5.0% of aoCRC patients. CONCLUSIONS Somatic and germline mutation profiles were similar for eoCRC and aoCRC patients and may not adequately explain differences in tumor behavior and age of disease onset.
Collapse
Affiliation(s)
| | - Qian Shi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Cathy Eng
- Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA
| | - Christopher Lieu
- Division of Medical Oncology, University of Colorado Health Cancer Center, Aurora, CO 80045, USA
| | - Thomas George
- Division of Hematology and Oncology, University of Florida, Gainesville, FL 32603, USA
| | | | | | - Binyam Yilma
- Tempus AI, Inc., Chicago, IL 60654, USA (E.A.T.)
| | | | | | - Amit Mahipal
- Department of Medical Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zhaohui Jin
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA;
| |
Collapse
|
4
|
Sorscher S. Health Care Equity and BRCA1/2 Testing. Health Equity 2025; 9:127-130. [PMID: 40123843 PMCID: PMC11848045 DOI: 10.1089/heq.2024.0167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2024] [Indexed: 03/25/2025] Open
Abstract
Although most cancers are sporadic, a significant proportion are related to inherited cancer-causing genes called pathogenic germline variants (PGVs). There are recommended measures for prevention and earlier diagnosis of cancers in patients identified as BRCA1 and BRCA2 PGV carriers, which are the most common cancer-predisposing PGVs. For example, published guidelines recommend that patients with BRCA1/2 PGVs undergo bilateral oophorectomies to prevent ovarian cancer and regular magnetic resonance imaging to screen for breast cancer. Also, those same measures are recommended for family members identified by cascade testing as BRCA1/2 carriers. Here, reports of the significant disparities between groups in which patients diagnosed with breast cancer are offered and undergo testing for BRCA1/2 PGVs are reviewed. Expanding the current standard of care guidelines for BRCA1/2 testing to all patients diagnosed with breast cancer and enacting the Cancer Moonshot 2.0 Initiative measures that should mitigate these disparities are discussed as well.
Collapse
|
5
|
Zhang X, Xu L, Cao Y, Ye P, Cheng Y, Lin X, Yi T, Wang P. Whole-Exome Sequencing Identifies Germline BLM Mutation in Ovarian Hepatoid Adenocarcinoma with Favorable Response to Niraparib and Anlotinib Combination Therapy-A Case Report and Literature Review. Int J Surg Pathol 2025; 33:236-248. [PMID: 39053024 DOI: 10.1177/10668969241260811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Hepatoid adenocarcinoma of the ovary represents a rare and malignant extrahepatic tumor that shares morphological and immunophenotypic similarities with hepatocellular carcinoma. Due to the ambiguous histomorphology and aggressive behavior, the diagnosis and management of hepatoid adenocarcinoma of the ovary present unique challenges. Here, we present a 67-year-old woman with massive ascites and disseminated peritoneal implants at initial diagnosis. She was treated with six cycles of neoadjuvant therapy (albumin-bound paclitaxel + nedaplatin + bevacizumab) and a debulking surgery, followed by eight cycles of postoperative adjuvant therapy (albumin-bound paclitaxel + carboplatin + bevacizumab). Elaborate pathology workup found significant involvement of angiogenesis in the tumor and confirmed the diagnosis via immunohistochemistry. Further molecular characterization of the tumor by whole-exome sequencing (WES) revealed a novel heterozygous germline mutation (NM_000057.2, c.1290_1291delinsATCAGGCCTCCATAG, p.Y430fs1) in gene BLM, likely pathogenic, suggesting a potential candidate for Poly (ADP-ribose) polymerase (PARP) inhibitors. For the maintenance therapy, she received a combination of the PARP inhibitor niraparib and the antiangiogenic anlotinib. As of now, the patient has achieved a partial response, with no apparent evidence of disease progression observed nearly 30 months. Our study sheds light on the WES-based profiling in rare cancers to screen for any treatable targets with otherwise no standard therapeutic options. The promising results with the niraparib-anlotinib combination suggest its potential as a maintenance therapy option for hepatoid adenocarcinoma of the ovary, which warrants validation in future larger cohort.
Collapse
Affiliation(s)
- Xiaofang Zhang
- Department of Gynecology and Obstetrics, West China Second University Hospital of Sichuan University, Chengdu, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, P.R. China
| | - Lian Xu
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, P.R. China
| | - Yidan Cao
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, P.R. China
| | - Pengfei Ye
- Department of Radiology, West China Second University Hospital of Sichuan University, Chengdu, P.R. China
| | - Yan Cheng
- Department of Radiology, West China Second University Hospital of Sichuan University, Chengdu, P.R. China
| | - Xiaojuan Lin
- Department of Gynecology and Obstetrics, West China Second University Hospital of Sichuan University, Chengdu, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, P.R. China
| | - Tianjin Yi
- Department of Gynecology and Obstetrics, West China Second University Hospital of Sichuan University, Chengdu, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, P.R. China
| | - Ping Wang
- Department of Gynecology and Obstetrics, West China Second University Hospital of Sichuan University, Chengdu, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, P.R. China
| |
Collapse
|
6
|
Nikanjam M, Kato S, Allen T, Sicklick JK, Kurzrock R. Novel clinical trial designs emerging from the molecular reclassification of cancer. CA Cancer J Clin 2025. [PMID: 39841128 DOI: 10.3322/caac.21880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
Next-generation sequencing has revealed the disruptive reality that advanced/metastatic cancers have complex and individually distinct genomic landscapes, necessitating a rethinking of treatment strategies and clinical trial designs. Indeed, the molecular reclassification of cancer suggests that it is the molecular underpinnings of the disease, rather than the tissue of origin, that mostly drives outcomes. Consequently, oncology clinical trials have evolved from standard phase 1, 2, and 3 tissue-specific studies; to tissue-specific, biomarker-driven trials; to tissue-agnostic trials untethered from histology (all drug-centered designs); and, ultimately, to patient-centered, N-of-1 precision medicine studies in which each patient receives a personalized, biomarker-matched therapy/combination of drugs. Innovative technologies beyond genomics, including those that address transcriptomics, immunomics, proteomics, functional impact, epigenetic changes, and metabolomics, are enabling further refinement and customization of therapy. Decentralized studies have the potential to improve access to trials and precision medicine approaches for underserved minorities. Evaluation of real-world data, assessment of patient-reported outcomes, use of registry protocols, interrogation of exceptional responders, and exploitation of synthetic arms have all contributed to personalized therapeutic approaches. With greater than 1 × 1012 potential patterns of genomic alterations and greater than 4.5 million possible three-drug combinations, the deployment of artificial intelligence/machine learning may be necessary for the optimization of individual therapy and, in the near future, also may permit the discovery of new treatments in real time.
Collapse
Affiliation(s)
- Mina Nikanjam
- Division of Hematology-Oncology, University of California San Diego, La Jolla, California, USA
- Moores Cancer Center, University of California San Diego Health, La Jolla, California, USA
| | - Shumei Kato
- Division of Hematology-Oncology, University of California San Diego, La Jolla, California, USA
- Moores Cancer Center, University of California San Diego Health, La Jolla, California, USA
| | | | - Jason K Sicklick
- Moores Cancer Center, University of California San Diego Health, La Jolla, California, USA
- Division of Surgical Oncology, Department of Surgery, University of California San Diego, San Diego, California, USA
- Department of Pharmacology, University of California San Diego, San Diego, California, USA
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin, USA
- Worldwide Innovative Networking in Personalized Cancer Medicine Consortium, Paris, France
| |
Collapse
|
7
|
Shore N, Nielsen SM, Esplin ED, Antonarakis ES, Barata PC, Beer TM, Beltran H, Bryce A, Cookson MS, Crawford ED, Dorff TB, George DJ, Heath EI, Helfand BT, Hussain M, Mckay RR, Morgans AK, Morris MJ, Paller CJ, Ross AE, Sartor O, Shen J, Sieber P, Smith MR, Wise DR, Armstrong AJ. Implementation of Universal Germline Genetic Testing Into Standard of Care for Patients With Prostate Cancer: The Time Is Now. JCO Oncol Pract 2024:OP2400626. [PMID: 39700441 DOI: 10.1200/op-24-00626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/25/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024] Open
Abstract
Indications for and implications of germline genetic testing (GGT) in patients with prostate cancer have expanded over the past decade, particularly related to precision therapies and management. GGT has become the standard of care for many cancers such as breast, ovarian, colorectal, pancreatic, and metastatic prostate cancer, and it is imperative that patients be offered timely and equitable access to testing as it can inform patient-physician shared decision making for management of the current cancer as well as anticipatory guidance for disease progression. Additionally, GGT guides screening for and prevention of secondary malignancies for the patient and cascade testing for at-risk family members. Here, we present data supporting the notion that clinicians should offer all patients with prostate cancer the opportunity to undergo comprehensive GGT for pathogenic germline variants known to be associated with familial cancer and/or known to have implications for treatment and management.
Collapse
Affiliation(s)
- Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC
| | - Sarah M Nielsen
- Labcorp Genetics Inc (formerly Invitae Corp), San Francisco, CA
| | - Edward D Esplin
- Labcorp Genetics Inc (formerly Invitae Corp), San Francisco, CA
| | | | | | - Tomasz M Beer
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Exact Sciences Corporation, Madison, WI
| | | | - Alan Bryce
- City of Hope Cancer Center, Goodyear, AZ
| | - Michael S Cookson
- Stephenson Cancer Center, OU Health, The University of Oklahoma, Oklahoma City, OK
| | | | | | | | | | - Brian T Helfand
- NorthShore University HealthSystem/Endeavor Health, Evanston, IL
| | - Maha Hussain
- Northwestern Feinberg School of Medicine, Chicago, IL
| | | | | | | | - Channing J Paller
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ashley E Ross
- Northwestern Feinberg School of Medicine, Chicago, IL
| | | | - John Shen
- UCLA David Geffen School of Medicine, Los Angeles, CA
| | | | | | - David R Wise
- Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | | |
Collapse
|
8
|
Mollica L, Quaquarini E, Schiepatti A, Travaglino E, Antoci F, Vanoli A, Arpa G, Biagi F, Locati LD. A small bowel adenocarcinoma harboring a DDR2 mutation in a celiac patient. Clin J Gastroenterol 2024; 17:1026-1032. [PMID: 39117782 DOI: 10.1007/s12328-024-02025-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024]
Abstract
We present the case of a 62-year-old man with a history of celiac disease and IgA deficiency, following a strict gluten-free diet that was admitted to our hospital for recurrent abdominal pain, fatigue and melena. Esophagogastroduodenoscopy and colonoscopy with biopsies were normal. A video-capsule endoscopy was performed and revealed a sub-stenosing, vegetating, and bleeding lesion in the first jejunal loop. He underwent laparotomic surgery with resection of the involved segment with loco-regional lymphadenectomy. The pathological report described a poorly differentiated adenocarcinoma of the jejunum, stage IIIA (pT3pN1). Analysis of next-generation sequencing (NGS) of DNA on the surgical sample revealed a likely pathogenetic variant in exon 15 of the DDR2 gene (c.2003G > A) and a TP53 non-frame-shift deletion (c.585_602del). Considering the risk of recurrence, he was candidate to 6 months of adjuvant chemotherapy with platinum salt and fluoropyrimidine. Thirty-eight months after the diagnosis, the patient is still disease free and in good clinical condition. This is the first described case of SBA with DDR2 mutation. Considering the limited therapeutic options beyond surgery for SBA, molecular analyses could become promising for the search for potential targetable alterations for treatments with new available drugs.
Collapse
Affiliation(s)
- Ludovica Mollica
- Medical Oncology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri, 10 27100, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Erica Quaquarini
- Medical Oncology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri, 10 27100, Pavia, Italy.
| | - Annalisa Schiepatti
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- Gastroenterology Unit of Pavia Institute, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Erica Travaglino
- Anatomic Pathology Unit, Fondazione IRCCS San Matteo Hospital, Pavia, Italy
| | - Francesca Antoci
- Anatomic Pathology Unit, Fondazione IRCCS San Matteo Hospital, Pavia, Italy
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Giovanni Arpa
- Anatomical Pathology Unit of Pavia Institute, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Federico Biagi
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- Gastroenterology Unit of Pavia Institute, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Laura Deborah Locati
- Medical Oncology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri, 10 27100, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| |
Collapse
|
9
|
Landry KK, DeSarno MJ, Kipnis L, Barquet Ramos F, Breen KM, Patton K, Morrissette A, Buehler RM, Ukaegbu C, Rohanizadegan M, Yurgelun MB, Syngal S, Rana HQ, Garber JE. Prevalence and Distribution of Unexpected Actionable Germline Pathogenic Variants Identified on Broad-Based Multigene Panel Testing Among Patients With Cancer. JCO Precis Oncol 2024; 8:e2400553. [PMID: 39666927 DOI: 10.1200/po-24-00553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/27/2024] [Accepted: 11/01/2024] [Indexed: 12/14/2024] Open
Abstract
PURPOSE In patients with a variety of malignancies undergoing multigene panel testing (MGPT), we examined the frequency of a pathogenic/likely pathogenic variant (PV) that would not have been predicted on the basis of the patient's personal and family history of cancer. METHODS This is a retrospective review of patients with cancer ascertained from a single academic cancer center who underwent broad-based MGPT of ≥20 cancer predisposition genes not selected on the basis of personal or family cancer history from 2015 to 2021. Low-penetrance variants and recessive inheritance genes were excluded. Deidentified pedigrees were analyzed to determine clinical suspicion of PV. RESULTS MGPT was performed on 10,975 patients with cancer: 1,134 (10.3%) were found to have ≥1 PV in a moderate or highly penetrant cancer susceptibility gene. Three hundred seven (2.8%) of the PVs were not predicted on the basis of patient's personal cancer history alone, and 192 (1.7%) remained unsuspected after patient's cancer diagnosis and review of family cancer histories were considered. Unexpected PVs accounted for 16.9% of the 1,134 patients with a moderate- or high-penetrance PV. Most frequent unexpected variants were MITF (n = 18), PMS2 (n = 18), ATM (n = 17), BRIP1 (n = 17), HOXB13 (n = 14), SDHA (n = 12), CHEK2 (n = 11), BRCA2 (n = 7), MSH6 (n = 7), SDHC (n = 7), PALB2 (n = 6), and TP53 (n = 6). Low-penetrance or recessive variants were found in 519 (4.7%) patients. Variants of uncertain significance were found in 3,775 (34.4%). CONCLUSION In patients with cancer, MGPT identified a rate of 1.7% PV in unexpected actionable cancer predisposition genes. Findings were more often unexpected (2.8%) when considering only the patient cancer history. These findings may justify consideration of broader MGPT panels in patients with cancer, given implications for subsequent surveillance, cascade testing, and treatment options dependent on specific findings.
Collapse
Affiliation(s)
- Kara K Landry
- Division of Hematology and Oncology, Department of Medicine, University of Vermont Medical Center, Burlington, VT
| | - Michael J DeSarno
- Department of Medical Biostatistics, Larner College of Medicine, University of Vermont, Burlington, VT
| | - Lindsay Kipnis
- Divison of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Farid Barquet Ramos
- Divison of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Katelyn M Breen
- Divison of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Kaleigh Patton
- Divison of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Audrey Morrissette
- Divison of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Ryan M Buehler
- Divison of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Chinedu Ukaegbu
- Divison of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Mersedeh Rohanizadegan
- Division of Translational Medicine and Human Genetics, University of Pennsylvania, Philadelphia, PA
| | - Matthew B Yurgelun
- Divison of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Sapna Syngal
- Divison of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Huma Q Rana
- Divison of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Judy E Garber
- Divison of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
10
|
Manirakiza AV, Baichoo S, Uwineza A, Dukundane D, Uwinkindi F, Ngendahayo E, Rubagumya F, Muhawenimana E, Nsabimana N, Nzeyimana I, Maniragaba T, Ntirenganya F, Rurangwa E, Mugenzi P, Mutamuliza J, Runanira D, Niyibizi BA, Rugengamanzi E, Besada J, Nielsen SM, Bucknor B, Nussbaum RL, Koeller D, Andrews C, Mutesa L, Fadelu T, Rebbeck TR. Germline sequence variation in cancer genes in Rwandan breast and prostate cancer cases. NPJ Genom Med 2024; 9:61. [PMID: 39582020 PMCID: PMC11586404 DOI: 10.1038/s41525-024-00446-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024] Open
Abstract
Cancer genetic data from Sub-Saharan African (SSA) are limited. Patients with female breast (fBC), male breast (mBC), and prostate cancer (PC) in Rwanda underwent germline genetic testing and counseling. Demographic and disease-specific information was collected. A multi-cancer gene panel was used to identify germline Pathogenic Variants (PV) and Variants of Uncertain Significance (VUS). 400 patients (201 with BC and 199 with PC) were consented and recruited to the study. Data was available for 342 patients: 180 with BC (175 women and 5 men) and 162 men with PC. PV were observed in 18.3% fBC, 4.3% PC, and 20% mBC. BRCA2 was the most common PV. Among non-PV carriers, 65% had ≥1 VUS: 31.8% in PC and 33.6% in BC (female and male). Our findings highlight the need for germline genetic testing and counseling in cancer management in SSA.
Collapse
Affiliation(s)
- Achille Vc Manirakiza
- Oncology Unit, Department of Medicine, King Faisal Hospital, Kigali, Rwanda.
- College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda.
| | - Shakuntala Baichoo
- Department of Digital Technologies, University of Mauritius, Réduit, Mauritius
| | - Annette Uwineza
- Centre for Human Genetics, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
- Kigali University Teaching Hospital, Kigali, Rwanda
| | - Damas Dukundane
- Oncology Unit, Department of Medicine, King Faisal Hospital, Kigali, Rwanda
| | | | - Edouard Ngendahayo
- Urology Unit, Department of Surgery, King Faisal Hospital, Kigali, Rwanda
| | - Fidel Rubagumya
- College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
- Rwanda Cancer Center, Rwanda Military Hospital, Kigali, Rwanda
| | | | | | | | | | - Faustin Ntirenganya
- College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
- Kigali University Teaching Hospital, Kigali, Rwanda
| | - Ephrem Rurangwa
- Rwanda Military Referral and Teaching Hospital, Kigali, Rwanda
| | | | | | | | | | | | | | | | | | - Robert L Nussbaum
- Invitae, Inc, San Francisco, CA, USA
- Department of Pediatrics, Division of Medical Genetics, University of California in San Francisco, San Francisco, CA, USA
| | | | | | - Leon Mutesa
- Centre for Human Genetics, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | - Temidayo Fadelu
- Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Timothy R Rebbeck
- Dana Farber Cancer Institute, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
11
|
Westphalen CB, Martins-Branco D, Beal JR, Cardone C, Coleman N, Schram AM, Halabi S, Michiels S, Yap C, André F, Bibeau F, Curigliano G, Garralda E, Kummar S, Kurzrock R, Limaye S, Loges S, Marabelle A, Marchió C, Mateo J, Rodon J, Spanic T, Pentheroudakis G, Subbiah V. The ESMO Tumour-Agnostic Classifier and Screener (ETAC-S): a tool for assessing tumour-agnostic potential of molecularly guided therapies and for steering drug development. Ann Oncol 2024; 35:936-953. [PMID: 39187421 DOI: 10.1016/j.annonc.2024.07.730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/19/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Advances in precision oncology led to approval of tumour-agnostic molecularly guided treatment options (MGTOs). The minimum requirements for claiming tumour-agnostic potential remain elusive. METHODS The European Society for Medical Oncology (ESMO) Precision Medicine Working Group (PMWG) coordinated a project to optimise tumour-agnostic drug development. International experts examined and summarised the publicly available data used for regulatory assessment of the tumour-agnostic indications approved by the US Food and Drug Administration and/or the European Medicines Agency as of December 2023. Different scenarios of minimum objective response rate (ORR), number of tumour types investigated, and number of evaluable patients per tumour type were assessed for developing a screening tool for tumour-agnostic potential. This tool was tested using the tumour-agnostic indications approved during the first half of 2024. A taxonomy for MGTOs and a framework for tumour-agnostic drug development were conceptualised. RESULTS Each tumour-agnostic indication had data establishing objective response in at least one out of five patients (ORR ≥ 20%) in two-thirds (≥4) of the investigated tumour types, with at least five evaluable patients in each tumour type. These minimum requirements were met by tested indications and may serve as a screening tool for tumour-agnostic potential, requiring further validation. We propose a conceptual taxonomy classifying MGTOs based on the therapeutic effect obtained by targeting a driver molecular aberration across tumours and its modulation by tumour-specific biology: tumour-agnostic, tumour-modulated, or tumour-restricted. The presence of biology-informed mechanistic rationale, early regulatory advice, and adequate trial design demonstrating signs of biology-driven tumour-agnostic activity, followed by confirmatory evidence, should be the principles for tumour-agnostic drug development. CONCLUSION The ESMO Tumour-Agnostic Classifier (ETAC) focuses on the interplay of targeted driver molecular aberration and tumour-specific biology modulating the therapeutic effect of MGTOs. We propose minimum requirements to screen for tumour-agnostic potential (ETAC-S) as part of tumour-agnostic drug development. Definition of ETAC cut-offs is warranted.
Collapse
Affiliation(s)
- C B Westphalen
- Comprehensive Cancer Center Munich & Department of Medicine III, University Hospital, LMU Munich, Munich; German Cancer Consortium (DKTK), partner site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - D Martins-Branco
- Scientific and Medical Division, European Society for Medical Oncology (ESMO), Lugano, Switzerland
| | - J R Beal
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - C Cardone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Naples, Italy
| | - N Coleman
- School of Medicine, Trinity College Dublin, Dublin; Medical Oncology Department, St. James's Hospital, Dublin; Trinity St. James's Cancer Institute, Dublin, Ireland
| | - A M Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City; Weill Cornell Medical College, New York City
| | - S Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham; Duke Cancer Institute, Duke University, Durham, USA
| | - S Michiels
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif, France
| | - C Yap
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - F André
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre
| | - F Bibeau
- Service d'Anatomie Pathologique, CHU Besançon, Université de Bourgogne Franche-Comté, Besançon, France
| | - G Curigliano
- Istituto Europeo di Oncologia, IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - E Garralda
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - S Kummar
- Division of Hematology and Medical Oncology, Department of Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland
| | - R Kurzrock
- Department of Medicine, Medical College of Wisconsin Cancer Center, Milwaukee, USA
| | - S Limaye
- Medical & Precision Oncology, Sir H. N. Reliance Foundation Hospital & Research Centre, Mumbai, India
| | - S Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), German Center for Lung Research (DZL), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - A Marabelle
- Drug Development Department (DITEP) and Laboratory for Translational Research in Immunotherapy (LRTI), Gustave Roussy, INSERM U1015 & CIC1428, Université Paris-Saclay, Villejuif, France
| | - C Marchió
- Department of Medical Sciences, University of Turin, Turin; Division of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - J Mateo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - J Rodon
- Department of Investigational Cancer Therapeutics, UT MD Anderson, Houston, USA
| | - T Spanic
- Europa Donna Slovenia, Ljubljana, Slovenia
| | - G Pentheroudakis
- Scientific and Medical Division, European Society for Medical Oncology (ESMO), Lugano, Switzerland
| | - V Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute (SCRI), Nashville, USA
| |
Collapse
|
12
|
Dinić J, Jovanović Stojanov S, Dragoj M, Grozdanić M, Podolski-Renić A, Pešić M. Cancer Patient-Derived Cell-Based Models: Applications and Challenges in Functional Precision Medicine. Life (Basel) 2024; 14:1142. [PMID: 39337925 PMCID: PMC11433531 DOI: 10.3390/life14091142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/22/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
The field of oncology has witnessed remarkable progress in personalized cancer therapy. Functional precision medicine has emerged as a promising avenue for achieving superior treatment outcomes by integrating omics profiling and sensitivity testing of patient-derived cancer cells. This review paper provides an in-depth analysis of the evolution of cancer-directed drugs, resistance mechanisms, and the role of functional precision medicine platforms in revolutionizing individualized treatment strategies. Using two-dimensional (2D) and three-dimensional (3D) cell cultures, patient-derived xenograft (PDX) models, and advanced functional assays has significantly improved our understanding of tumor behavior and drug response. This progress will lead to identifying more effective treatments for more patients. Considering the limited eligibility of patients based on a genome-targeted approach for receiving targeted therapy, functional precision medicine provides unprecedented opportunities for customizing medical interventions according to individual patient traits and individual drug responses. This review delineates the current landscape, explores limitations, and presents future perspectives to inspire ongoing advancements in functional precision medicine for personalized cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11108 Belgrade, Serbia; (J.D.); (S.J.S.); (M.D.); (M.G.); (A.P.-R.)
| |
Collapse
|
13
|
Istl AC, Lawton S, Kamaraju S, Stolley M, Petroll AE, Cortina CS. Tumors, Treatments, and Trust: Cancer Characteristics, Outcomes, and Screening Uptake in Transgender and Gender-Diverse Patients. Ann Surg Oncol 2024; 31:5560-5569. [PMID: 38861206 PMCID: PMC11309885 DOI: 10.1245/s10434-024-15319-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/04/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND More than 2.5 million adults in the United States identify as transgender or gender-diverse (TGD), but little data exist on cancer screening and care for this population. We examined cancer characteristics, screening adherence, genetic testing, and provider inclusive language for TGD patients with cancer. METHODS This single institution retrospective cohort study identified TGD patients with cancer between 2000 and 2022. Demographic, clinicopathological, treatment, and screening data were collected, as well as data on gender-affirming care (GAC) and use of patients' personal pronouns in medical records. Descriptive statistics and regression analyses were used to report outcomes. RESULTS Sixty unique patients with 69 cancer diagnoses were included: 63.3% were transgender women, 21.7% transgender men, 6.7% nonbinary, and 8.3% were genderqueer. Sixty-five percent had a family history of cancer. Only 46.2% of those who met genetic testing criteria were referred. On review of recommended cancer screening, colorectal screening had the greatest uptake (62%), followed by breast (48.3%), lung (35.7%), cervical (33.3%), and prostate (32%); 8.5% of cancers were diagnosed on screening. Individuals with Medicare had reduced odds of screening uptake (OR 0.07, 95% CI 0.01-0.58) versus private insurance. With respect to GAC, 73.3% used gender-affirming hormone therapy and 41% had gender-affirming surgery. After initiating GAC and asserting personal pronouns, 75% were referred to by incorrect name/pronouns in provider documentation. CONCLUSIONS Our TGD cancer patient cohort had low rates of disease-specific cancer screening and inadequate genetic referrals. Many providers did not use appropriate patient names/pronouns. Provider and patient interventions are needed to ensure inclusive preventative and oncologic care for this marginalized population.
Collapse
Affiliation(s)
- Alexandra C Istl
- Division of Surgical Oncology, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Samuel Lawton
- Division of Surgical Oncology, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sailaja Kamaraju
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
| | - Melinda Stolley
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
| | - Andrew E Petroll
- Division of Infectious Disease, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Froedtert and the Medical College of Wisconsin Inclusion Health Clinic, Milwaukee, WI, USA
| | - Chandler S Cortina
- Division of Surgical Oncology, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
| |
Collapse
|
14
|
Marconi G, Rondoni M, Zannetti BA, Zacheo I, Nappi D, Mattei A, Rocchi S, Lanza F. Novel insights and therapeutic approaches in secondary AML. Front Oncol 2024; 14:1400461. [PMID: 39135995 PMCID: PMC11317385 DOI: 10.3389/fonc.2024.1400461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/05/2024] [Indexed: 08/15/2024] Open
Abstract
Secondary acute myeloid leukemia (sAML) presents as a complex and multifaceted ensemble of disorders, positioning itself as both a challenge and an intriguing frontier within hematologic oncology. Its origins are diverse, stemming from antecedent hematologic conditions, germline predisposing mutations, or the sequelae of cytotoxic therapies, and its development is driven by intricate genetic and epigenetic modifications. This complexity necessitates a diverse array of therapeutic strategies, each meticulously tailored to address the distinctive challenges sAML introduces. Such strategies require a personalized approach, considering the variegated clinical backgrounds of patients and the inherent intricacies of the disease. Allogeneic stem cell transplantation stands as a cornerstone, offering the potential for curative outcomes. This is complemented by the emergence of innovative treatments such as CPX-351, venetoclax, and glasdegib, which have demonstrated promising results in enhancing prognosis. The evolving landscape of sAML treatment underscores the importance of continued research and innovation in the field, aiming not only to improve patient outcomes but also to deepen our understanding of the disease's biological underpinnings, thereby illuminating pathways toward more effective and individualized therapies.
Collapse
Affiliation(s)
- Giovanni Marconi
- Hematology Unit and Romagna Transplant Network, Hospital of Ravenna, University of Bologna, Ravenna, Italy
| | - Michela Rondoni
- Hematology Unit and Romagna Transplant Network, Hospital of Ravenna, Ravenna, Italy
| | | | - Irene Zacheo
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Davide Nappi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Agnese Mattei
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Serena Rocchi
- Hematology Unit and Romagna Transplant Network, Hospital of Ravenna, Ravenna, Italy
| | - Francesco Lanza
- Hematology Unit and Romagna Transplant Network, Hospital of Ravenna, University of Bologna, Ravenna, Italy
| |
Collapse
|
15
|
Mohindroo C, Baydogan S, Agarwal P, Wright RD, Prakash LR, Mork ME, Klein AP, Laheru DA, Maxwell JE, Katz MHG, Dasari A, Kim MP, He J, McAllister F, De Jesus-Acosta A. Germline Testing Identifies Pathogenic/Likely Pathogenic Variants in Patients with Pancreatic Neuroendocrine Tumors. Cancer Prev Res (Phila) 2024; 17:335-342. [PMID: 38662083 DOI: 10.1158/1940-6207.capr-23-0483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/10/2024] [Accepted: 04/23/2024] [Indexed: 04/26/2024]
Abstract
Ten percent of pancreatic neuroendocrine tumors (pNET) are related to inherited syndromes (MEN1, MEN4, VHL, NF1, and TSC). Growing evidence suggests that clinically sporadic pNETs can also harbor germline pathogenic variants. In this study, we report the prevalence of pathologic/likely pathologic (P/LP) germline variants in a high-risk cohort and an unselected cohort. We collected clinical data of patients with pNETs seen at MD Anderson Cancer Center and Johns Hopkins Hospital. The high-risk cohort included (n = 132) patients seen at MD Anderson Cancer Center who underwent germline testing for high-risk criteria (early onset, personal or family history of cancer, and syndromic features) between 2013 and 2019. The unselected cohort included (n = 106) patients seen at Johns Hopkins Hospital who underwent germline testing following their diagnosis of pNETs between 2020 and 2022. In the high-risk cohort (n = 132), 33% (n = 44) had P/LP variants. The majority of the patients had P/LP variants in MEN1 56% (n = 25), followed by DNA repair pathways 18% (n = 8), and 7% (n = 3) in MSH2 (Lynch syndrome). Patients with P/LP were younger (45 vs. 50 years; P = 0.002). In the unselected cohort (n = 106), 21% (n = 22) had P/LP. The majority were noted in DNA repair pathways 40% (n = 9) and MEN1 36% (n = 8). Multifocal tumors correlated with the presence of P/LP (P = 0.0035). MEN1 germline P/LP variants correlated with younger age (40 vs. 56 years; P = 0.0012), presence of multifocal tumors (P < 0.0001), and World Health Organization grade 1 histology (P = 0.0078). P/LP variants are prevalent in patients with clinically sporadic pNET irrespective of high-risk features. The findings support upfront universal germline testing in all patients with pNET. Prevention Relevance: Here, we present germline data from the largest reported cohort of patients with pNET (n = 238), comprising both a high-risk cohort and an unselected cohort. In both cohorts, we identify a high number of P/LPs, including those in the DNA repair pathway. Our findings support universal germline testing in patients with pNET.
Collapse
Affiliation(s)
- Chirayu Mohindroo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Seyda Baydogan
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Parul Agarwal
- Department of Oncology, Abramson Cancer Center at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robin D Wright
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laura R Prakash
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maureen E Mork
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alison P Klein
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel A Laheru
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jessica E Maxwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael P Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jin He
- Department of Surgical Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ana De Jesus-Acosta
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
Subbiah V, Horgan D, Subbiah IM. A Vision for Democratizing Next-Generation Oncology Clinical Trials. Cancer Discov 2024; 14:579-584. [PMID: 38571427 DOI: 10.1158/2159-8290.cd-24-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
SUMMARY Revolutionary advancements in oncology have transformed lives, but the clinical trials ecosystem encounters challenges, including restricted access to innovative therapies and a lack of diversity in participant representation. A vision emerges for democratized, globally accessible oncology trials, necessitating collaboration among researchers, clinicians, patients, and policymakers to shift from converting complex, exclusive trials into a dynamic, inclusive force against cancer.
Collapse
Affiliation(s)
- Vivek Subbiah
- Sarah Cannon Research Institute, Nashville, Tennessee
| | - Denis Horgan
- European Alliance for Personalized Medicine, Brussels, Belgium
| | - Ishwaria M Subbiah
- Sarah Cannon Research Institute, Nashville, Tennessee
- The US Oncology Network, The Woodlands, Texas
| |
Collapse
|
17
|
Pollard RD, Wilkerson MD, Rajagopal PS. Identification of germline population variants misclassified as cancer-associated somatic variants. Front Med (Lausanne) 2024; 11:1361317. [PMID: 38572163 PMCID: PMC10987807 DOI: 10.3389/fmed.2024.1361317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 02/28/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction Databases used for clinical interpretation in oncology rely on genetic data derived primarily from patients of European ancestry, leading to biases in cancer genetics research and clinical practice. One practical issue that arises in this context is the potential misclassification of multi-ancestral population variants as tumor-associated because they are not represented in reference genomes against which tumor sequencing data is aligned. Methods To systematically find misclassified variants, we compared somatic variants in census genes from the Catalogue of Somatic Mutations in Cancer (COSMIC) V99 with multi-ancestral population variants from the Genome Aggregation Databases' Linkage Disequilibrium (GnomAD). By comparing genomic coordinates, reference, and alternate alleles, we could identify misclassified variants in genes associated with cancer. Results We found 192 of 208 genes in COSMIC's cancer-associated census genes (92.31%) to be associated with variant misclassifications. Among the 1,906,732 variants in COSMIC, 6,957 variants (0.36%) aligned with normal population variants in GnomAD, concerning for misclassification. The African / African American ancestral population included the greatest number of misclassified variants and also had the greatest number of unique misclassified variants. Conclusion The direct, systematic comparison of variants from COSMIC for co-occurrence in GnomAD supports a more accurate interpretation of tumor sequencing data and reduces bias related to genomic ancestry.
Collapse
Affiliation(s)
- Rebecca D. Pollard
- Maret School, Washington, DC, United States
- Metis Foundation, San Antonio, TX, United States
| | - Matthew D. Wilkerson
- Center for Military Precision Health, Uniformed Services University, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Padma Sheila Rajagopal
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
18
|
Kamaraju S, Conroy M, Harris A, Georgen M, Min H, Powell M, Kurzrock R. Challenges to genetic testing for germline mutations associated with breast cancer among African Americans. Cancer Treat Rev 2024; 124:102695. [PMID: 38325071 DOI: 10.1016/j.ctrv.2024.102695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
Inequities in preventive cancer screening, diagnosis, treatment, and inferior cancer outcomes continue to pose challenges across the cancer continuum. While the exact reasons for these inferior outcomes are unknown, multiple barriers to various domains of social determinants of health (SDOH) play a vital role, leading to inequities in cancer care. These include barriers to transportation, housing, and food insecurities, contributing to delays in preventive screening and treatment. Furthermore, aggressive biologies also exist across various racial profiles with accompanying germline mutations. For example, African Americans (AAs) have a higher incidence of triple-negative breast cancer subtype and a high prevalence of BRCA1/2 gene mutations, increasing the risk of multiple cancers, warranting high-risk screening for these populations. Unfortunately, other barriers, such as financial insecurities, low health literacy rates, and lack of awareness, lead to delays in cancer screening and genetic testing, even with available high-risk screening and risk reduction procedures. In addition, physicians receive minimal interdisciplinary training to address genetic assessment, interpretation of the results, and almost no additional training in addressing the unique needs of racial minorities, leading to suboptimal delivery of genetic assessment provision resources among AAs. In this review, we discuss the confluence of factors and barriers limiting genetic testing among AAs and highlight the prevalence of germline mutations associated with increased risk of breast cancer among AAs, reflecting the need for multi-panel germline testing as well as education regarding hereditary cancer risks in underserved minorities.
Collapse
Affiliation(s)
- S Kamaraju
- Medical College of Wisconsin, Milwaukee, WI, USA; Department of Medicine, Division of Hematology-Oncology, Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA.
| | - M Conroy
- Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| | - A Harris
- Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| | - M Georgen
- Medical College of Wisconsin, Milwaukee, WI, USA; Department of Medicine, Division of Hematology-Oncology, Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| | - H Min
- Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| | - M Powell
- Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| | - R Kurzrock
- Medical College of Wisconsin, Milwaukee, WI, USA; Department of Medicine, Division of Hematology-Oncology, Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| |
Collapse
|
19
|
Aryal B, Bizhanova Z, Joseph EA, Yin Y, Wagner PL, Dalton E, LaFramboise WA, Bartlett DL, Allen CJ. Navigating Precision Oncology: Insights from an Integrated Clinical Data and Biobank Repository Initiative across a Network Cancer Program. Cancers (Basel) 2024; 16:760. [PMID: 38398150 PMCID: PMC10886699 DOI: 10.3390/cancers16040760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Advancing cancer treatment relies on the rapid translation of new scientific discoveries to patient care. To facilitate this, an oncology biobank and data repository program, also referred to as the "Moonshot" program, was launched in 2021 within the Integrated Network Cancer Program of the Allegheny Health Network. A clinical data program (CDP) and biospecimen repository were established, and patient data and blood and tissue samples have been collected prospectively. To date, the study has accrued 2920 patients, predominantly female (61%) and Caucasian (90%), with a mean age of 64 ± 13 years. The most common cancer sites were the endometrium/uterus (12%), lung/bronchus (12%), breast (11%), and colon/rectum (11%). Of patients diagnosed with cancer, 34% were diagnosed at stage I, 25% at stage II, 26% at stage III, and 15% at stage IV. The CDP is designed to support our initiative in advancing personalized cancer research by providing a comprehensive array of patient data, encompassing demographic characteristics, diagnostic details, and treatment responses. The "Moonshot" initiative aims to predict therapy responses and clinical outcomes through cancer-related biomarkers. The CDP facilitates this initiative by fostering data sharing, enabling comparative analyses, and informing the development of novel diagnostic and therapeutic methods.
Collapse
Affiliation(s)
- Bibek Aryal
- Allegheny Singer Research Institute, Allegheny Health Network, Pittsburgh, PA 15212, USA; (B.A.); (Z.B.); (E.A.J.); (Y.Y.)
| | - Zhadyra Bizhanova
- Allegheny Singer Research Institute, Allegheny Health Network, Pittsburgh, PA 15212, USA; (B.A.); (Z.B.); (E.A.J.); (Y.Y.)
| | - Edward A. Joseph
- Allegheny Singer Research Institute, Allegheny Health Network, Pittsburgh, PA 15212, USA; (B.A.); (Z.B.); (E.A.J.); (Y.Y.)
| | - Yue Yin
- Allegheny Singer Research Institute, Allegheny Health Network, Pittsburgh, PA 15212, USA; (B.A.); (Z.B.); (E.A.J.); (Y.Y.)
| | - Patrick L. Wagner
- Division of Surgical Oncology, Institute of Surgery, Allegheny Health Network, Pittsburgh, PA 15212, USA;
| | | | | | - David L. Bartlett
- Allegheny Health Network Cancer Institute, Pittsburgh, PA 15212, USA;
| | - Casey J. Allen
- Division of Surgical Oncology, Institute of Surgery, Allegheny Health Network, Pittsburgh, PA 15212, USA;
| |
Collapse
|
20
|
Riedl JM, Moik F, Esterl T, Kostmann SM, Gerger A, Jost PJ. Molecular diagnostics tailoring personalized cancer therapy-an oncologist's view. Virchows Arch 2024; 484:169-179. [PMID: 37982847 PMCID: PMC10948510 DOI: 10.1007/s00428-023-03702-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/27/2023] [Accepted: 11/04/2023] [Indexed: 11/21/2023]
Abstract
Medical oncology is rapidly evolving with the implementation of personalized, targeted therapies. Advances in molecular diagnostics and the biologic understanding of cancer pathophysiology led to the identification of specific genetic alterations as drivers of cancer progression. Further, improvements in drug development enable the direct interference with these pathways, which allow tailoring personalized treatments based on a distinct molecular characterization of tumors. Thereby, we are currently experiencing a paradigm-shift in the treatment of cancers towards cancer-type agnostic, molecularly targeted, personalized therapies. However, this concept has several important hurdles and limitations to overcome to ultimately increase the proportion of patients benefitting from the precision oncology approach. These include the assessment of clinical relevancy of identified alterations, capturing and interpreting levels of heterogeneity based on intra-tumoral or time-dependent molecular evolution, and challenges in the practical implementation of precision oncology in routine clinical care. In the present review, we summarize the current state of cancer-agnostic precision oncology, discuss the concept of molecular tumor boards, and consider current limitations of personalized cancer therapy. Further, we provide an outlook towards potential future developments including the implementation of functionality assessments of identified genetic alterations and the broader use of liquid biopsies in order to obtain more comprehensive and longitudinal genetic information that might guide personalized cancer therapy in the future.
Collapse
Affiliation(s)
- Jakob M Riedl
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florian Moik
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Tamara Esterl
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Sarah M Kostmann
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Philipp J Jost
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
- Medical Department III for Haematology and Oncology, School of Medicine, Technical University of Munich, Munich, Germany.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
21
|
Hofman P, Berezowska S, Kazdal D, Mograbi B, Ilié M, Stenzinger A, Hofman V. Current challenges and practical aspects of molecular pathology for non-small cell lung cancers. Virchows Arch 2024; 484:233-246. [PMID: 37801103 PMCID: PMC10948551 DOI: 10.1007/s00428-023-03651-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023]
Abstract
The continuing evolution of treatment options in thoracic oncology requires the pathologist to regularly update diagnostic algorithms for management of tumor samples. It is essential to decide on the best way to use tissue biopsies, cytological samples, as well as liquid biopsies to identify the different mandatory predictive biomarkers of lung cancers in a short turnaround time. However, biological resources and laboratory member workforce are limited and may be not sufficient for the increased complexity of molecular pathological analyses and for complementary translational research development. In this context, the surgical pathologist is the only one who makes the decisions whether or not to send specimens to immunohistochemical and molecular pathology platforms. Moreover, the pathologist can rapidly contact the oncologist to obtain a new tissue biopsy and/or a liquid biopsy if he/she considers that the biological material is not sufficient in quantity or quality for assessment of predictive biomarkers. Inadequate control of algorithms and sampling workflow may lead to false negative, inconclusive, and incomplete findings, resulting in inappropriate choice of therapeutic strategy and potentially poor outcome for patients. International guidelines for lung cancer treatment are based on the results of the expression of different proteins and on genomic alterations. These guidelines have been established taking into consideration the best practices to be set up in clinical and molecular pathology laboratories. This review addresses the current predictive biomarkers and algorithms for use in thoracic oncology molecular pathology as well as the central role of the pathologist, notably in the molecular tumor board and her/his participation in the treatment decision-making. The perspectives in this setting will be discussed.
Collapse
Affiliation(s)
- Paul Hofman
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France.
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France.
| | - Sabina Berezowska
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Daniel Kazdal
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Baharia Mograbi
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France
| | - Marius Ilié
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France
| | - Albrecht Stenzinger
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Véronique Hofman
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France
| |
Collapse
|
22
|
Koster R, Schipper LJ, Giesbertz NAA, van Beek D, Mendeville M, Samsom KG, Rosenberg EH, Hogervorst FBL, Roepman P, Boelens MC, Bosch LJW, van den Berg JG, Meijer GA, Voest EE, Cuppen E, Ruijs MWG, van Wezel T, van der Kolk L, Monkhorst K. Impact of genetic counseling strategy on diagnostic yield and workload for genome-sequencing-based tumor diagnostics. Genet Med 2024; 26:101032. [PMID: 38006283 DOI: 10.1016/j.gim.2023.101032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/17/2023] [Accepted: 11/19/2023] [Indexed: 11/26/2023] Open
Abstract
PURPOSE Genome sequencing (GS) enables comprehensive molecular analysis of tumors and identification of hereditary cancer predisposition. According to guidelines, directly determining pathogenic germline variants (PGVs) requires pretest genetic counseling, which is cost-ineffective. Referral for genetic counseling based on tumor variants alone could miss relevant PGVs and/or result in unnecessary referrals. METHODS We validated GS for detection of germline variants and simulated 3 strategies using paired tumor-normal GS data of 937 metastatic patients. In strategy-1, genetic counseling before tumor testing allowed direct PGV analysis. In strategy-2 and -3, germline testing and referral for post-test genetic counseling is based on tumor variants using Dutch (strategy-2) or Europen Society for Medical Oncology (ESMO) Precision Medicine Working Group (strategy-3) guidelines. RESULTS In strategy-1, PGVs would be detected in 50 patients (number-needed-to counsel; NTC = 18.7). In strategy-2, 86 patients would have been referred for genetic counseling and 43 would have PGVs (NTC = 2). In strategy-3, 94 patients would have been referred for genetic counseling and 32 would have PGVs (NTC = 2.9). Hence, 43 and 62 patients, respectively, were unnecessarily referred based on a somatic variant. CONCLUSION Both post-tumor test counseling strategies (2 and 3) had significantly lower NTC, and strategy-2 had the highest PGV yield. Combining pre-tumor test mainstreaming and post-tumor test counseling may maximize the clinically relevant PGV yield and minimize unnecessary referrals.
Collapse
Affiliation(s)
- Roelof Koster
- The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Luuk J Schipper
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | - Kris G Samsom
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Paul Roepman
- Hartwig Medical Foundation, Amsterdam, The Netherlands
| | | | - Linda J W Bosch
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Gerrit A Meijer
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Emile E Voest
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Edwin Cuppen
- Hartwig Medical Foundation, Amsterdam, The Netherlands
| | | | - Tom van Wezel
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Kim Monkhorst
- The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
23
|
Kim J, Vaksman Z, Egolf LE, Kaufman R, Evans JP, Conkrite KL, Danesh A, Lopez G, Randall MP, Dent MH, Farra LM, Menghani NL, Dymek M, Desai H, Hausler R, Hicks B, Guidry Auvil JM, Gerhard DS, Hakonarson H, Maxwell KN, Cole KA, Pugh TJ, Bosse KR, Khan J, Wei JS, Maris JM, Stewart DR, Diskin SJ. Germline pathogenic variants in neuroblastoma patients are enriched in BARD1 and predict worse survival. J Natl Cancer Inst 2024; 116:149-159. [PMID: 37688579 PMCID: PMC10777667 DOI: 10.1093/jnci/djad183] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/02/2023] [Accepted: 08/25/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Neuroblastoma is an embryonal cancer of the developing sympathetic nervous system. The genetic contribution of rare pathogenic or likely pathogenic germline variants in patients without a family history remains unclear. METHODS Germline DNA sequencing was performed on 786 neuroblastoma patients. The frequency of rare cancer predisposition gene pathogenic or likely pathogenic variants in patients was compared with 2 cancer-free control cohorts. Matched tumor DNA sequencing was evaluated for second hits, and germline DNA array data from 5585 neuroblastoma patients and 23 505 cancer-free control children were analyzed to identify rare germline copy number variants. Patients with germline pathogenic or likely pathogenic variants were compared with those without to test for association with clinical characteristics, tumor features, and survival. RESULTS We observed 116 pathogenic or likely pathogenic variants involving 13.9% (109 of 786) of neuroblastoma patients, representing a statistically significant excess burden compared with cancer-free participants (odds ratio [OR] = 1.60, 95% confidence interval [CI] = 1.27 to 2.00). BARD1 harbored the most statistically significant enrichment of pathogenic or likely pathogenic variants (OR = 32.30, 95% CI = 6.44 to 310.35). Rare germline copy number variants disrupting BARD1 were identified in patients but absent in cancer-free participants (OR = 29.47, 95% CI = 1.52 to 570.70). Patients harboring a germline pathogenic or likely pathogenic variant had a worse overall survival compared with those without (P = 8.6 x 10-3). CONCLUSIONS BARD1 is an important neuroblastoma predisposition gene harboring both common and rare germline pathogenic or likely pathogenic variations. The presence of any germline pathogenic or likely pathogenic variant in a cancer predisposition gene was independently predictive of worse overall survival. As centers move toward paired tumor-normal sequencing at diagnosis, efforts should be made to centralize data and provide an infrastructure to support cooperative longitudinal prospective studies of germline pathogenic variation.
Collapse
Affiliation(s)
- Jung Kim
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Zalman Vaksman
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laura E Egolf
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca Kaufman
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - J Perry Evans
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Karina L Conkrite
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Arnavaz Danesh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, ON, Canada
| | - Gonzalo Lopez
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michael P Randall
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Maiah H Dent
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lance M Farra
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Neil L Menghani
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Malwina Dymek
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Heena Desai
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Hausler
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Belynda Hicks
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | | | - Daniela S Gerhard
- Office of Cancer Genomics, National Cancer Institute, Bethesda, MD, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kara N Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristina A Cole
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Kristopher R Bosse
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Javed Khan
- Oncogenomics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jun S Wei
- Oncogenomics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - John M Maris
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Sharon J Diskin
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Papadopoulou E, Rigas G, Fountzilas E, Boutis A, Giassas S, Mitsimponas N, Daliani D, Ziogas DC, Liontos M, Ramfidis V, Christophilakis C, Matthaios D, Floros T, Florou-Chatzigiannidou C, Agiannitopoulos K, Meintani A, Tsantikidi A, Katseli A, Potska K, Tsaousis G, Metaxa-Mariatou V, Nasioulas G. Microsatellite Instability Is Insufficiently Used as a Biomarker for Lynch Syndrome Testing in Clinical Practice. JCO Precis Oncol 2024; 8:e2300332. [PMID: 38271656 PMCID: PMC10830089 DOI: 10.1200/po.23.00332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/06/2023] [Accepted: 11/15/2023] [Indexed: 01/27/2024] Open
Abstract
PURPOSE The pan-cancer presence of microsatellite instability (MSI)-positive tumors demonstrates its clinical utility as an agnostic biomarker for identifying immunotherapy-eligible patients. Additionally, MSI is a hallmark of Lynch syndrome (LS), the most prevalent cancer susceptibility syndrome among patients with colorectal and endometrial cancer. Therefore, MSI-high results should inform germline genetic testing for cancer-predisposing genes. However, in clinical practice, such analysis is frequently disregarded. METHODS A next-generation sequencing (NGS)-based technique was used for MSI analysis in 4,553 patients with various tumor types. Upon request, somatic BRAF gene analysis was conducted. In addition, hereditary testing of cancer-associated genes was performed in MSI-high cases using a capture-based NGS protocol. MLH1 promoter methylation analysis was conducted retrospectively in patients with colorectal and endometrial cancer to further investigate the origin of MSI at the tumor level. RESULTS The MSI positivity rate for the entire cohort was 5.27%. Endometrial, gastric, colorectal, urinary tract, and prostate cancers showed the highest proportion of MSI-high cases (15.69%, 8.54%, 7.40%, 4.55%, and 3.19%, respectively). A minority of 45 patients (22.73%) among the MSI-high cases underwent germline testing to determine whether the mismatch repair pathway deficiency was inherited. 24.44% of those who performed the genetic test carried a pathogenic variant in an LS-associated gene. Three MSI-high individuals had non-LS gene alterations, including BRCA1, BRCA2, and CDKN2A pathogenic variants, indicating the presence of non-LS-associated gene alterations among MSI-high patients. CONCLUSION Although MSI analysis is routinely performed in clinical practice, as many as 77% of MSI-high patients do not undergo LS genetic testing, despite international guidelines strongly recommending it. BRAF and MLH1 methylation analysis could shed light on the somatic origin of MSI in 42.50% of the MSI-high patients; however, MLH1 analysis is barely ever requested in clinical practice.
Collapse
Affiliation(s)
| | - George Rigas
- Medical Oncology Unit, General Hospital of Volos, Volos, Greece
| | - Elena Fountzilas
- Second Department of Medical Oncology, Euromedica General Clinic, Thessaloniki, Greece
| | - Anastasios Boutis
- First Department of Clinical Oncology, Theagenio Hospital, Thessaloniki, Greece
| | - Stylianos Giassas
- Second Oncology Clinic IASO, General Maternity and Gynecology Clinic, Athens, Greece
| | | | - Danai Daliani
- Department of Medical Oncology, Euroclinic, Athens, Greece
| | - Dimitrios C Ziogas
- First Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | - Michalis Liontos
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, “Alexandra” General Hospital of Athens, Athens, Greece
| | | | | | | | - Theofanis Floros
- Oncology Department, Athens Naval and Veterans Hospital, Athens, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Subbiah V, Burris HA, Kurzrock R. Revolutionizing cancer drug development: Harnessing the potential of basket trials. Cancer 2024; 130:186-200. [PMID: 37934000 DOI: 10.1002/cncr.35085] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 11/08/2023]
Abstract
The landscape of cancer therapy has been transformed by advances in clinical next-generation sequencing, genomically targeted therapies, and immunotherapies. Well designed clinical trials and efficient clinical trial conduct are crucial for advancing our understanding of cancer, improving patient outcomes, and identifying personalized treatments. Basket trials have emerged as one of the efficient modern clinical trial designs that evaluate the efficacy of these therapies across multiple cancer types based on specific molecular alterations or biomarkers, irrespective of histology or anatomic location. This review delves into the evolution of basket trials in cancer drug development, highlighting their potential prospects and current obstacles. The design of basket trials involves screening patients for specific molecular alterations or biomarkers and enrolling them in the trial to receive the targeted therapy under investigation. Statistical considerations play a crucial role in the design, analysis, and interpretation of basket trials. Several notable examples of basket trials that have led to US Food and Drug Administration approval for uncommon molecular alterations (e.g., NTRK fusions, BRAF mutations, RET and FGFR1 alterations) are discussed, including LOXO-TRK (ClinicalTrials.gov identifier NCT02122913)/SCOUT (ClinicalTrials.gov identifier NCT02637687)/NAVIGATE (ClinicalTrials.gov identifier NCT02576431)/STARTRK (ClinicalTrials.gov identifiers NT02097810, NT02568267), VE-BASKET (ClinicalTrials.gov identifier NCT01524978), ROAR Basket (ClinicalTrials.gov identifier NCT02034110), LIBRETTO-001 (ClinicalTrials.gov identifier NCT03157128), ARROW (ClinicalTrials.gov identifier NCT03037385), FIGHT-203 (ClinicalTrials.gov identifier NCT03011372), and the National Cancer Institute-Molecular Analysis for Therapy Choice trial (ClinicalTrials.gov identifier NCT02465060). Basket trials have the potential to revolutionize cancer treatment by identifying effective therapies for patients based on specific molecular alterations or biomarkers rather than traditional histology-based approaches. PLAIN LANGUAGE SUMMARY: To gain more knowledge about cancer, improve patient outcomes, and discover personalized treatments, it is crucial to conduct clinical trials efficiently. One effective type of clinical trial is called a basket trial. In basket trials, new treatments are tested on various types of cancer, regardless of their location in the body; instead, researchers focus on specific abnormalities in the cancer cells. Basket trials offer hope that we can find personalized treatments that are more effective for each individual battling cancer.
Collapse
Grants
- Boehringer Ingelheim, Debiopharm, Foundation Medicine, Genentech, Grifols, Guardant, Incyte, Konica Minolta, Medimmune, Merck Serono, Omniseq, Pfizer, Sequenom, Takeda, and TopAlliance and from the NCI
Collapse
Affiliation(s)
- Vivek Subbiah
- Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | | | - Razelle Kurzrock
- Department of Medicine, Medical College of Wisconsin Cancer Center and Genome Sciences and Precision Medicine Center, Milwaukee, Wisconsin, USA
- WIN Consortium, Paris, France
| |
Collapse
|
26
|
Moyers JT, Subbiah V. TRKing sarcomas for precision medicine: Seek, and ye shall find! Cancer 2023; 129:3694-3696. [PMID: 37769119 DOI: 10.1002/cncr.34941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Plain language summary
The way in which we treat different types of cancer has changed a lot because of genome‐driven precision oncology. This approach uses information from a person’s genes to personalize their treatment.
A recent study conducted by Kummar and colleagues confirms that neurotrophic receptor tyrosine kinase (NTRK) inhibitors are beneficial in treating sarcomas with NTRK fusions.
This study adds to the growing evidence that genomics, which is the study of genes and their functions, can help us to identify effective treatments even for rare targets such as NTRK fusions.
This emphasizes the importance of using advanced DNA sequencing techniques in analyzing the genetic makeup of all sarcomas.
Collapse
Affiliation(s)
- Justin T Moyers
- Angeles Clinic and Research Institute, Cedars-Sinai Affiliate, Los Angeles, California, USA
| | - Vivek Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute, Nashville, Tennessee, USA
| |
Collapse
|
27
|
Shore N, Gazi M, Pieczonka C, Heron S, Modh R, Cahn D, Belkoff LH, Berger A, Mazzarella B, Veys J, Idom C, Morris D, Jayram G, Engelman A, Bukkapatnam R, Dato P, Bevan-Thomas R, Cornell R, Wise DR, Hardwick MK, Hernandez RD, Rojahn S, Layman P, Hatchell KE, Heald B, Nussbaum RL, Nielsen SM, Esplin ED. Efficacy of National Comprehensive Cancer Network Guidelines in Identifying Pathogenic Germline Variants Among Unselected Patients with Prostate Cancer: The PROCLAIM Trial. Eur Urol Oncol 2023; 6:477-483. [PMID: 37574391 DOI: 10.1016/j.euo.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/07/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND Prostate cancer (PCa) patients with pathogenic/likely pathogenic germline variants (PGVs) in cancer predisposition genes may be eligible for U.S. Food and Drug Administration-approved targeted therapies, clinical trials, or enhanced screening. Studies suggest that eligible patients are missing genetics-informed care due to restrictive testing criteria. OBJECTIVE To establish the prevalence of actionable PGVs among prospectively accrued, unselected PCa patients, stratified by their guideline eligibility. DESIGN, SETTING, AND PARTICIPANTS Consecutive, unselected PCa patients were enrolled at 15 sites in the USA from October 2019 to August 2021, and had multigene cancer panel testing. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Correlates between the prevalence of PGVs and clinician-reported demographic and clinical characteristics were examined. RESULTS AND LIMITATIONS Among 958 patients (median [quartiles] age at diagnosis 65 [60, 71] yr), 627 (65%) had low- or intermediate-risk disease (grade group 1, 2, or 3). A total of 77 PGVs in 17 genes were identified in 74 patients (7.7%, 95% confidence interval [CI] 6.2-9.6%). No significant difference was found in the prevalence of PGVs among patients who met the 2019 National Comprehensive Cancer Network Prostate criteria (8.8%, 43/486, 95% CI 6.6-12%) versus those who did not (6.6%, 31/472, 95% CI 4.6-9.2%; odds ratio 1.38, 95% CI 0.85-2.23), indicating that these criteria would miss 42% of patients (31/74, 95% CI 31-53%) with PGVs. The criteria were less effective at predicting PGVs in patients from under-represented populations. Most PGVs (81%, 60/74) were potentially clinically actionable. Limitations include the inability to stratify analyses based on individual ethnicity due to low numbers of non-White patients with PGVs. CONCLUSIONS Our results indicate that almost half of PCa patients with PGVs are missed by current testing guidelines. Comprehensive germline genetic testing should be offered to all patients with PCa. PATIENT SUMMARY One in 13 patients with prostate cancer carries an inherited variant that may be actionable for the patient's current care or prevention of future cancer, and could benefit from expanded testing criteria.
Collapse
Affiliation(s)
- Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA.
| | - Mukaram Gazi
- University Urology Associates of New Jersey, Hamilton, NJ, USA
| | | | - Sean Heron
- Advanced Urology Institute, St. Petersburg, FL, USA
| | - Rishi Modh
- Advanced Urology Institute, St. Petersburg, FL, USA
| | | | | | - Aaron Berger
- Associated Urological Specialists, Chicago Ridge, IL, USA
| | | | | | | | | | | | | | | | - Paul Dato
- Genesis Healthcare Partners, San Diego, CA, USA
| | | | | | - David R Wise
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | | | - Ryan D Hernandez
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | | - Robert L Nussbaum
- Invitae Corporation, San Francisco, CA, USA; Volunteer Faculty, University of California San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
28
|
Seligson ND, Kolesar JM, Alam B, Baker L, Lamba JK, Fridley BL, Salahudeen AA, Hertz DL, Hicks JK. Integrating pharmacogenomic testing into paired germline and somatic genomic testing in patients with cancer. Pharmacogenomics 2023; 24:731-738. [PMID: 37702060 DOI: 10.2217/pgs-2023-0125] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023] Open
Abstract
Precision medicine has revolutionized clinical care for patients with cancer through the development of targeted therapy, identification of inherited cancer predisposition syndromes and the use of pharmacogenetics to optimize pharmacotherapy for anticancer drugs and supportive care medications. While germline (patient) and somatic (tumor) genomic testing have evolved separately, recent interest in paired germline/somatic testing has led to an increase in integrated genomic testing workflows. However, paired germline/somatic testing has generally lacked the incorporation of germline pharmacogenomics. Integrating pharmacogenomics into paired germline/somatic genomic testing would be an efficient method for increasing access to pharmacogenomic testing. In this perspective, the authors argue for the benefits of implementing a comprehensive approach integrating somatic and germline testing that is inclusive of pharmacogenomics in clinical practice.
Collapse
Affiliation(s)
- Nathan D Seligson
- Department of Pharmacotherapy & Translational Research, The University of Florida, Jacksonville, FL 32209, USA
- Center for Pharmacogenomics & Translational Research, Nemours Children's Health, Jacksonville, FL 32207, USA
| | - Jill M Kolesar
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
- Department of Pharmacy Practice & Science, University of Kentucky, Lexington, KY 40536, USA
| | - Benish Alam
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA
| | - Laura Baker
- Nemours Center for Cancer & Blood Disorders, Nemours Children's Health, Wilmington, DE 19803, USA
| | - Jatinder K Lamba
- Department of Pharmacotherapy & Translational Research, The University of Florida, Gainesville, FL 32611, USA
| | - Brooke L Fridley
- Department of Biostatistics & Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Ameen A Salahudeen
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Tempus Labs Inc., Chicago, IL 60654, USA
| | - Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA
| | - J Kevin Hicks
- Department of Individualized Cancer Management, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
29
|
Wen H, Xu Q, Sheng X, Li H, Wang X, Wu X. Prevalence and Landscape of Pathogenic or Likely Pathogenic Germline Variants and Their Association With Somatic Phenotype in Unselected Chinese Patients With Gynecologic Cancers. JAMA Netw Open 2023; 6:e2326437. [PMID: 37523182 PMCID: PMC10391307 DOI: 10.1001/jamanetworkopen.2023.26437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/01/2023] Open
Abstract
Importance Understanding germline and somatic status in patients with gynecologic cancers could improve risk assessment and guide therapeutic decision-making. Objective To evaluate the prevalence and landscape of germline pathogenic or likely pathogenic (P/LP) variants and explore whether these variants are associated with somatic phenotypes and cancer risk in unselected patients with gynecologic cancers. Design, Setting, and Participants This cross-sectional study retrospectively enrolled unselected patients in China with a gynecologic cancer, including ovarian, cervical, and endometrial, who underwent tumor-normal sequencing using a 520-gene panel from October 1, 2017, through May 31, 2021. Exposure Germline variants in gynecologic cancers. Main Outcomes and Measures The P/LP germline variant rates in 62 cancer predisposition genes were assessed using descriptive statistics. The associations of P/LP variant status with age, somatic profiles, and cancer risk were also investigated using the Fisher exact test or Student t test. Results A total of 1610 women (median [IQR] age, 54 [47-62] years; 1201 [74.6%] with stage III-IV disease) were included (945 with ovarian cancer, 307 with endometrial cancer, and 358 with cervical cancer). The prevalence of patients with P/LP variants was 20.5% (194 of 945) for ovarian cancer, 13.4% (41 of 307) for endometrial cancer, and 6.4% (23 of 358) for cervical cancer; 95.1% of the germline findings (n = 252) were potentially actionable, mainly in homologous recombination repair (HRR) and mismatch repair genes. Chinese patients with endometrial cancer had a higher rate of P/LP variants than a White population from The Cancer Genome Atlas (42 of 307 [13.7%] vs 24 of 367 [6.5%]; P = .003). In endometrial and cervical cancers, the prevalence of P/LP variants was 12.7% (30 of 237) and 4.8% (13 of 270), respectively, in patients diagnosed at age 45 years or older and increased to 25.0% (9 of 36; P = .09) and 12.0% (10 of 83; P = .04), respectively, for those with an onset age of less than 45 years. Mismatch repair P/LP variants were associated with a younger age at onset for ovarian cancer (46 vs 54 years; P = .02) and endometrial cancer (48 vs 57 years; P < .001), while HRR P/LP variants were associated with a younger age at onset for cervical cancer (46 vs 52 years; P = .04). Carriers of HRR P/LP variants had more prevalent somatic TP53 variants and less common somatic variants in oncogenic driver genes vs noncarriers. BRCA1/2 P/LP variants were also associated with moderate risks for endometrial and cervical cancer. Conclusions and Relevance This study delineates the landscape of germline P/LP variants in Chinese women with gynecologic cancers. The findings highlight the hereditary factor in cervical cancer that has long been neglected and suggest the importance of next-generation sequencing-based genetic testing with a large gene panel for gynecologic cancers.
Collapse
Affiliation(s)
- Hao Wen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qin Xu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xiujie Sheng
- Department of Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huawen Li
- Department of Gynecology, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|