3551
|
Cho D, Gor B, Hwang H, Wang X, Hernandez M, Jones LA, Frost J, Roberson P, Pettaway CA. A Community-Based Prostate Cancer Screening and Education Program for Asian American Men in Medically Underserved Communities. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:415. [PMID: 38673327 PMCID: PMC11050265 DOI: 10.3390/ijerph21040415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024]
Abstract
This study analyzed data from a community-based prostate cancer (PCa) education and screening program (Prostate Outreach Project; POP) to enhance PCa-related knowledge among medically underserved Asian American men. It also examined PCa screening history, clinical abnormalities based on prostate-specific antigen (PSA) tests and digital rectal examination (DRE) results, and follow-up and PCa diagnosis rates. Participants-521 Asian men (251 Vietnamese, 142 Chinese, and 128 South Asians)-were offered PCa screening using PSA tests and/or DRE and an educational session on PCa. Of these men, 277 completed PCa-related knowledge surveys before and after viewing an educational video. Significant between-group differences in PCa-related knowledge were found at pre-assessment (p < 0.001) but not at post-assessment (p = 0.11), at which time all groups showed improved PCa-related knowledge. Most participants (77.9%) had never received PCa screening, but Vietnamese men had the lowest previous screening rate (17.3%). Chinese men had elevated PSA values and the highest abnormal DRE rates. Of the 125 men with abnormal screening outcomes, only 15.2% had adequate follow-up. Of the 144 men diagnosed with PCa in POP, 11.1% were Asians (seven Chinese, six Vietnamese, and three South Asian). Despite the ethnic heterogeneity among Asian men, a community outreach program may successfully enhance their PCa-related knowledge.
Collapse
Affiliation(s)
- Dalnim Cho
- Department of Health Disparities Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.C.); (B.G.)
| | - Beverly Gor
- Department of Health Disparities Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.C.); (B.G.)
| | - Hyunsoo Hwang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mike Hernandez
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lovell A. Jones
- Department of Health Disparities Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.C.); (B.G.)
| | - Jacqueline Frost
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pamela Roberson
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Curtis A. Pettaway
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
3552
|
Baek SW, Leem SH. A Multi-Gene Signature of Non-Muscle-Invasive Bladder Cancer Identifies Patients Who Respond to Immunotherapies Including Bacillus Calmette-Guérin and Immune Checkpoint Inhibitors. Int J Mol Sci 2024; 25:3800. [PMID: 38612609 PMCID: PMC11012101 DOI: 10.3390/ijms25073800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Approximately 75% of bladder cancer cases originate as non-muscle-invasive bladder cancer (NMIBC). Despite initial diagnosis, NMIBC commonly recurs, with up to 45% advancing to muscle-invasive bladder cancer (MIBC) and metastatic disease. Treatment for high-risk NMIBC typically includes procedures like transurethral resection and, depending on recurrence risk, intravesical chemotherapy or immunotherapy such as Bacillus Calmette-Guérin (BCG). However, persistent shortages of BCG necessitate alternative first-line treatments. We aim to use a multi-gene signature in high-risk NMIBC patients to determine whether patients may benefit from immune checkpoint inhibitors (ICIs) as an alternative to BCG and to evaluate their clinical utility. The multi-gene signature obtained from the three independent NMIBC cohorts was applied to stratify the UROMOL2016 cohort (n = 476) using consensus clustering. Each subtype was distinguished by biological pathway analysis. Validation analysis using a machine learning algorithm was performed in six independent cohorts including the BRS (n = 283) cohort treated with BCG and the IMvigor210 (n = 298) clinical trials treated with PD-L1 inhibitors. Based on consensus cluster analysis, NMIBC patients in the UROMOL2016 cohort were classified into three classes exhibiting distinguished characteristics, including DNA damage repair (DDR). Survival analysis showed that the NMIBC-DDR class had the highest rates of disease progression (progression-free survival, p = 0.002 by log-rank test) in the UROMOL cohort and benefited from BCG and ICIs (respectively, p = 0.02 and p = 0.03 by log-rank test). This study suggests that the multi-gene signature may have a role in identifying high-risk NMIBC patients and improving the responsiveness of ICIs. Additionally, we propose immunotherapy as a new first-line treatment for patients with high-risk NMIBC because of the shortage of BCG supply. It is important to help more patients prioritize cancer immunotherapy.
Collapse
Affiliation(s)
- Seung-Woo Baek
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea;
| | - Sun-Hee Leem
- Department of Biomedical Sciences, Dong-A University, Busan 49315, Republic of Korea
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| |
Collapse
|
3553
|
Loomans-Kropp HA. The utility of liquid biopsy-based methylation biomarkers for colorectal cancer detection. Front Oncol 2024; 14:1351514. [PMID: 38595823 PMCID: PMC11002156 DOI: 10.3389/fonc.2024.1351514] [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: 12/07/2023] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent cancers and the second leading cause of cancer-related deaths in the United States. It is also one of the few cancers with established screening guidelines, however these methods have significant patient burden (e.g., time, invasive). In recent years, the development of liquid biopsy-based screening methods for biomarker detection have emerged as alternatives to traditional screening. Methylation biomarkers are of particular interest, and these markers can be identified and measured on circulating tumor and cell-free DNA. This perspective summarizes the current state of CRC screening and the potential integration of DNA methylation markers into liquid biopsy-based techniques. Finally, I discuss limitations to these methods and strategies for improvement. The continued development and implementation of liquid biopsy-based cancer screening approaches may provide an acceptable alternative to individuals unwilling to be screened by traditional methods.
Collapse
Affiliation(s)
- Holli A. Loomans-Kropp
- Cancer Control Program, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, United States
- Division of Cancer Prevention and Control, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
3554
|
Tang C, Jiang ST, Li CX, Jia XF, Yang WL. The Effect of Salvianolic Acid A on Tumor-Associated Macrophage Polarization and Its Mechanisms in the Tumor Microenvironment of Triple-Negative Breast Cancer. Molecules 2024; 29:1469. [PMID: 38611749 PMCID: PMC11013304 DOI: 10.3390/molecules29071469] [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: 12/22/2023] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, with a high degree of malignancy and poor prognosis. Tumor-associated macrophages (TAMs) have been identified as significant contributors to the growth and metastasis of TNBC through the secretion of various growth factors and chemokines. Salvianolic acid A (SAA) has been shown to have anti-cancer activities. However, the potential activity of SAA on re-polarized TAMs remains unclear. As there is a correlation between the TAMs and TNBC, this study investigates the effect of SAA on TAMs in the TNBC microenvironment. For that purpose, M2 TAM polarization was induced by two kinds of TNBC-conditioned medium (TNBC-TCM) in the absence or presence of SAA. The gene and protein expression of TAM markers were analyzed by qPCR, FCM, IF, ELISA, and Western blot. The protein expression levels of ERK and p-ERK in M2-like TAMs were analyzed by Western blot. The migration and invasion properties of M2-like TAMs were analyzed by Transwell assays. Here, we demonstrated that SAA increased the expression levels of CD86, IL-1β, and iNOS in M2-like TAMs and, conversely, decreased the expression levels of Arg-1 and CD206. Moreover, SAA inhibited the migration and invasion properties of M2-like TAMs effectively and decreased the protein expression of TGF-β1 and p-ERK in a concentration-dependent manner, as well as TGF-β1 gene expression and secretion. Our current findings for the first time demonstrated that SAA inhibits macrophage polarization to M2-like TAMs by inhibiting the ERK pathway and promotes M2-like TAM re-polarization to the M1 TAMs, which may exert its anti-tumor effect by regulating M1/M2 TAM polarization. These findings highlight SAA as a potential regulator of M2 TAMs and the possibility of utilizing SAA to reprogram M2 TAMs offers promising insights for the clinical management of TNBC.
Collapse
Affiliation(s)
- Chao Tang
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (C.T.); (S.-T.J.); (C.-X.L.); (X.-F.J.)
| | - Shi-Ting Jiang
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (C.T.); (S.-T.J.); (C.-X.L.); (X.-F.J.)
| | - Cheng-Xia Li
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (C.T.); (S.-T.J.); (C.-X.L.); (X.-F.J.)
| | - Xiao-Fang Jia
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (C.T.); (S.-T.J.); (C.-X.L.); (X.-F.J.)
| | - Wen-Li Yang
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (C.T.); (S.-T.J.); (C.-X.L.); (X.-F.J.)
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
3555
|
Hussain S, Guo Y, Huo Y, Shi J, Hou Y. Regulation of cancer progression by CK2: an emerging therapeutic target. Med Oncol 2024; 41:94. [PMID: 38526625 DOI: 10.1007/s12032-024-02316-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/28/2024] [Indexed: 03/27/2024]
Abstract
Casein kinase II (CK2) is an enzyme with pleiotropic kinase activity that catalyzes the phosphorylation of lots of substrates, including STAT3, p53, JAK2, PTEN, RELA, and AKT, leading to the regulation of diabetes, cardiovascular diseases, angiogenesis, and tumor progression. CK2 is observed to have high expression in multiple types of cancer, which is associated with poor prognosis. CK2 holds significant importance in the intricate network of pathways involved in promoting cell proliferation, invasion, migration, apoptosis, and tumor growth by multiple pathways such as JAK2/STAT3, PI3K/AKT, ATF4/p21, and HSP90/Cdc37. In addition to the regulation of cancer progression, increasing evidence suggests that CK2 could regulate tumor immune responses by affecting immune cell activity in the tumor microenvironment resulting in the promotion of tumor immune escape. Therefore, inhibition of CK2 is initially proposed as a pivotal candidate for cancer treatment. In this review, we discussed the role of CK2 in cancer progression and tumor therapy.
Collapse
Affiliation(s)
- Shakeel Hussain
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yilei Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yu Huo
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Juanjuan Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yongzhong Hou
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
3556
|
Teng T, Shi H, Fan Y, Guo P, Zhang J, Qiu X, Feng J, Huang H. Metabolic responses to the occurrence and chemotherapy of pancreatic cancer: biomarker identification and prognosis prediction. Sci Rep 2024; 14:6938. [PMID: 38521793 PMCID: PMC10960848 DOI: 10.1038/s41598-024-56737-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024] Open
Abstract
As the most malignant tumor, the prognosis of pancreatic cancer is not ideal even in the small number of patients who can undergo radical surgery. As a highly heterogeneous tumor, chemotherapy resistance is a major factor leading to decreased efficacy and postoperative recurrence of pancreatic cancer. In this study, nuclear magnetic resonance (NMR)-based metabolomics was applied to identify serum metabolic characteristics of pancreatic ductal adenocarcinoma (PDAC) and screen the potential biomarkers for its diagnosis. Metabolic changes of patients with different CA19-9 levels during postoperative chemotherapy were also monitored and compared to identify the differential metabolites that may affect the efficacy of chemotherapy. Finally, 19 potential serum biomarkers were screened to serve the diagnosis of PDAC, and significant metabolic differences between the two CA19-9 stratifications of PDAC were involved in energy metabolism, lipid metabolism, amino acid metabolism, and citric acid metabolism. Enrichment analysis of metabolic pathways revealed six shared pathways by PDAC and chemotherapy such as alanine, aspartate and glutamate metabolism, arginine biosynthesis, glutamine and glutamate metabolism, citrate cycle, pyruvate metabolism, and glycogolysis/gluconeogeneis. The similarity between the metabolic characteristics of PDAC and the metabolic responses to chemotherapy provided a reference for clinical prediction of benefits of postoperative chemotherapy in PDAC patients.
Collapse
Affiliation(s)
- Tianhong Teng
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Han Shi
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yanying Fan
- Fuzhou Children Hospital of Fujian Province, Fuzhou, Fujian, China
| | - Pengfei Guo
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Jin Zhang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xinyu Qiu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jianghua Feng
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China.
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
3557
|
Jiang S, Gao L, Li J, Zhang F, Zhang Y, Liu J. N6-methyladenosine-modified circ_0000337 sustains bortezomib resistance in multiple myeloma by regulating DNA repair. Front Cell Dev Biol 2024; 12:1383232. [PMID: 38586304 PMCID: PMC10995360 DOI: 10.3389/fcell.2024.1383232] [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: 02/07/2024] [Accepted: 03/12/2024] [Indexed: 04/09/2024] Open
Abstract
Studies have shown that bortezomib resistance in multiple myeloma (MM) is mediated by the abnormalities of various molecules and microenvironments. Exploring these resistance mechanisms will improve the therapeutic efficacy of bortezomib. In this study, bone marrow tissues from three patients with MM, both sensitive and resistant to bortezomib, were collected for circRNA high-throughput sequencing analysis. The relationship between circ_0000337, miR-98-5p, and target gene DNA2 was analyzed by luciferase detection and verified by RT-qPCR. We first found that circ_0000337 was significantly upregulated in bortezomib-resistant MM tissues and cells, and overexpression of circ_0000337 could promote bortezomib resistance in MM cells. circ_0000337 may act as a miR-98-5p sponge to upregulate DNA2 expression, regulate DNA damage repair, and induce bortezomib resistance. Furthermore, it was determined that the increased circ_0000337 level in bortezomib-resistant cells was due to an increased N6-methyladenosine (m6A) level, resulting in enhanced RNA stability. In conclusion, the m6A level of circ_0000337 and its regulation may be a new and potential therapeutic target for overcoming bortezomib resistance in MM.
Collapse
Affiliation(s)
- Siyi Jiang
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Lili Gao
- Jinan Hospital of Integrated Chinese and Western Medicine, Jinan, China
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Fangrong Zhang
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yanan Zhang
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jing Liu
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
3558
|
Owosho AA, DeColibus KA, Okhuaihesuyi O, Levy LC. Prophylactic Use of Pentoxifylline and Tocopherol for Prevention of Osteoradionecrosis of the Jaw after Dental Extraction in Post-Radiated Oral and Oropharyngeal Cancer Patients: An Initial Case Series. Dent J (Basel) 2024; 12:83. [PMID: 38667995 PMCID: PMC11049290 DOI: 10.3390/dj12040083] [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: 02/16/2024] [Revised: 03/13/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Osteoradionecrosis of the jaw is a morbid complication of radiotherapy in patients with oral and oropharyngeal cancers that may be precipitated by dental extractions. Pentoxifylline and tocopherol (PENTO) has been utilized in the management of osteoradionecrosis and as prophylaxis for post-radiated head and neck oncology patients requiring an invasive dental procedure. This observational study aims to report the outcome of the prophylactic use of PENTO in the prevention of osteoradionecrosis of the jaw after dental extractions in post-radiated oral and oropharyngeal cancer patients and to review the current literature on this topic. Four post-radiated oral and oropharyngeal oncology patients were referred to the dental oncology clinic of the University Dental Practice, University of Tennessee Health Sciences Center for dental extractions. All four patients were prescribed pentoxifylline 400 mg BID (twice a day) and tocopherol 400 IU BID (oral tablets) for 2 weeks before extraction(s) and for 6 weeks after extraction(s). All patients were followed up every week after the second week post-extraction if feasible until the extraction site(s) healed (covered by mucosa). The assessment endpoint was defined as 6 weeks post-extraction with the outcomes assessed as using four categories determined by the area of exposed bone: complete healing (complete mucosal coverage of extraction site); partial healing (reduction in size of extraction site); no change; and progression (increase in size of the extraction site). At the assessment endpoint, all patients had complete healing of all extraction sites. The ORN rate at the patient level (0/4) and individual tooth level (0/8) was 0%. All patients tolerated the PENTO medications and no adverse effects from the use of these medications were reported. This limited study in addition to the other reviewed studies estimates the rate of ORN at the patient level as 3.2% (14/436) for post-radiated head and neck oncology patients after dental extractions/invasive oral procedures. In conclusion, this PENTO regimen can reduce/prevent the incidence of ORN in post-radiated head and neck oncology patients. This safe and cost-effective protocol (PENTO regimen) should be further evaluated as prophylaxis for post-radiated head and neck oncology patients requiring an invasive dental procedure. We recommend large prospective studies to be carried out to further validate these findings.
Collapse
Affiliation(s)
- Adepitan A. Owosho
- Department of Diagnostic Sciences, College of Dentistry/Department of Otolaryngology—Head & Neck Surgery, College of Medicine, The University of Tennessee Health Sciences Center, 875 Union Avenue, Memphis, TN 38163, USA
| | - Katherine A. DeColibus
- Department of Diagnostic Sciences, College of Dentistry, The University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Osariemen Okhuaihesuyi
- Missouri School of Dentistry and Oral Health, A.T. Still University, Kirksville, MO 63501, USA
| | - Layne C. Levy
- Advanced Education in General Dentistry, College of Dentistry, The University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| |
Collapse
|
3559
|
Lou E, Xiu J, Baca Y, Saeed A, Prakash A, Gholami S, Subramanian S, Starr TK, Fontana E, Pandey R, Lenz HJ, Shields AF, Nabhan C, Oberley M, Seeber A, El-Deiry W. Differential landscape of immune evasion in oncogenic RAS-driven primary and metastatic colorectal cancers. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200786. [PMID: 38596288 PMCID: PMC10963927 DOI: 10.1016/j.omton.2024.200786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 04/11/2024]
Abstract
Oncogenic drivers such as KRAS extensively modulate the tumor inflammatory microenvironment (TIME) of colorectal cancer (CRC). The influence of KRAS on modulating immune cell composition remains unclear. The objective of this study was to identify signatures of infiltrative immune cells and distinctive patterns that differ between RAS wild-type (WT) and oncogenic mutant (MT) CRC that explain immune evasion in MT tumors. A total of 7,801 CRC specimens were analyzed using next-generation DNA sequencing, whole-exome sequencing, and/or whole transcriptome sequencing. Deficiency of mismatch repair (dMMR)/microsatellite instability (MSI) and tumor mutation burden (TMB) were also assessed. KRAS mutations were present in 48% of CRC, similarly distributed in patients younger than vs. 50 years and older. In microsatellite stable (MSS) KRAS MT tumors, composition of the TIME included higher neutrophil infiltration and lower infiltration of B cells. MSI-H/dMMR was significantly more prevalent in RAS WT (9.1%) than in KRAS MT (2.9%) CRC. In MSS CRC, TMB-high cases were significantly higher in RAS MT (3.1%) than in RAS WT (2.1%) tumors. KRAS and NRAS mutations are associated with increased neutrophil infiltration, with codon-specific differences. These results demonstrate significant differences in the TIME of RAS mutant CRC that match previous reports of immunoevasive characteristics of such tumors.
Collapse
Affiliation(s)
- Emil Lou
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Anwaar Saeed
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ajay Prakash
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Timothy K. Starr
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, MN, USA
| | - Elisa Fontana
- Drug Development Unit, Sarah Cannon Research Institute UK, Marylebone, London, UK
| | - Ritu Pandey
- Arizona Cancer Center, Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anthony F. Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | | | | | - Andreas Seeber
- Department of Internal Medicine V (Hematology and Oncology), Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Wafik El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Legorreta Cancer Center, Brown University, Providence, RI, USA
| |
Collapse
|
3560
|
Liu R, Huang X, Yang S, Du W, Chen X, Li H. Discovery of an independent poor-prognosis subtype associated with tertiary lymphoid structures in breast cancer. Front Immunol 2024; 15:1364506. [PMID: 38571938 PMCID: PMC10987760 DOI: 10.3389/fimmu.2024.1364506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction Tertiary lymphoid structures (TLSs) are ectopic lymphoid formations that arise in non-lymphoid tissues due to chronic inflammation. The pivotal function of TLSs in regulating tumor invasion and metastasis has been established across several cancers, such as lung cancer, liver cancer, and melanoma, with a positive correlation between increased TLS presence and improved prognosis. Nevertheless, the current research about the clinical significance of TLSs in breast cancer remains limited. Methods In our investigation, we discovered TLS-critical genes that may impact the prognosis of breast cancer patients, and categorized breast cancer into three distinct subtypes based on critical gene expression profiles, each exhibiting substantial differences in prognosis (p = 0.0046, log-rank test), with Cluster 1 having the best prognosis, followed by Cluster 2, and Cluster 3 having the worst prognosis. We explored the impact of the heterogeneity of these subtypes on patient prognosis, the differences in the molecular mechanism, and their responses to drug therapy and immunotherapy. In addition, we designed a machine learning-based classification model, unveiling highly consistent prognostic distinctions in several externally independent cohorts. Results A notable marker gene CXCL13 was identified in Cluster 3, potentially pivotal in enhancing patient prognosis. At the single-cell resolution, we delved into the adverse prognosis of Cluster 3, observing an enhanced interaction between fibroblasts, myeloid cells, and basal cells, influencing patient prognosis. Furthermore, we identified several significantly upregulated genes (CD46, JAG1, IL6, and IL6R) that may positively correlate with cancer cells' survival and invasive capabilities in this subtype. Discussion Our study is a robust foundation for precision medicine and personalized therapy, presenting a novel perspective for the contemporary classification of breast cancer.
Collapse
Affiliation(s)
- Ruiqi Liu
- School of Mathematics and Computer Science, Yunnan Minzu University, Kunming, China
| | - Xiaoqian Huang
- School of Mathematics and Computer Science, Yunnan Minzu University, Kunming, China
| | - Shiwei Yang
- School of Mathematics and Computer Science, Yunnan Minzu University, Kunming, China
| | - Wenbo Du
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiaozhou Chen
- School of Mathematics and Computer Science, Yunnan Minzu University, Kunming, China
| | - Huamei Li
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
3561
|
Situ J, Yang Y, Zhang L, Yan H, Cheng Y. Integration of O 2-economised tumour-targeted photosensitive magnetic nanomaterials in the diagnosis and therapy of gastric cancer. RSC Adv 2024; 14:9920-9932. [PMID: 38528931 PMCID: PMC10961965 DOI: 10.1039/d4ra00497c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/09/2024] [Indexed: 03/27/2024] Open
Abstract
Hypoxia in the tumour microenvironment is a major limiting factor in photodynamic therapy. The present study employed a novel O2-economised photosensitizer, ACSN, to effectively curtail oxygen consumption by impeding the aerobic respiration of tumour cells, thereby increasing the reactive oxygen species (ROS) production in photodynamic therapy. To enhance the efficacy of photodynamic therapy, the active targeting peptide iRGD was employed to facilitate drug accumulation in the tumour tissue. Therefore, we constructed a targeted drug platform, ACSN/Fe3O4@MSNs-iRGD, that integrates diagnosis and treatment. The drug exhibited excellent active targeting ability towards gastric cancer MGC-803 cells and can efficiently penetrate the mitochondria upon cellular internalisation. The photosensitizer ACSN, released from the drug, effectively suppressed mitochondrial aerobic respiration to conserve oxygen and exhibited robust ROS production upon laser excitation. The core-shell structure comprises Fe3O4, which offers excellent T2 dark contrast for real-time tumour monitoring through MRI imaging. By incorporating excellent photodynamic therapy and MRI imaging capabilities, this drug can serve as an effective platform for the integration of tumour diagnosis and treatment, thus addressing the limitations associated with conventional tumour therapies. It is anticipated that this approach will soon be clinically translated.
Collapse
Affiliation(s)
- JinRong Situ
- College of Fisheries and Life Science of Shanghai Ocean University Shanghai 201306 China
- Shanghai Jiao Tong University Affiliated Sixth People's Hosptial China
| | - Yingying Yang
- College of Fisheries and Life Science of Shanghai Ocean University Shanghai 201306 China
- Shanghai Jiao Tong University Affiliated Sixth People's Hosptial China
| | - Lingle Zhang
- College of Fisheries and Life Science of Shanghai Ocean University Shanghai 201306 China
- Shanghai Jiao Tong University Affiliated Sixth People's Hosptial China
| | - Hongzhang Yan
- College of Fisheries and Life Science of Shanghai Ocean University Shanghai 201306 China
- Shanghai Jiao Tong University Affiliated Sixth People's Hosptial China
| | - Yingsheng Cheng
- College of Fisheries and Life Science of Shanghai Ocean University Shanghai 201306 China
- Shanghai Jiao Tong University Affiliated Sixth People's Hosptial China
- Tongji Hospital Affiliated to Tongji University Shanghai China
| |
Collapse
|
3562
|
Pan J, Zhang J, Lin J, Cai Y, Zhao Z. Constructing lactylation-related genes prognostic model to effectively predict the disease-free survival and treatment responsiveness in prostate cancer based on machine learning. Front Genet 2024; 15:1343140. [PMID: 38566813 PMCID: PMC10985269 DOI: 10.3389/fgene.2024.1343140] [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: 11/27/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Background: Prostate cancer (PCa) is one of the most common malignancies in men with a poor prognosis. It is therefore of great clinical importance to find reliable prognostic indicators for PCa. Many studies have revealed the pivotal role of protein lactylation in tumor development and progression. This research aims to analyze the effect of lactylation-related genes on PCa prognosis. Methods: By downloading mRNA-Seq data of TCGA PCa, we obtained the differential genes related to lactylation in PCa. Five machine learning algorithms were used to screen for lactylation-related key genes for PCa, then the five overlapping key genes were used to construct a survival prognostic model by lasso cox regression analysis. Furthermore, the relationships between the model and related pathways, tumor mutation and immune cell subpopulations, and drug sensitivity were explored. Moreover, two risk groups were established according to the risk score calculated by the five lactylation-related genes (LRGs). Subsequently, a nomogram scoring system was established to predict disease-free survival (DFS) of patients by combining clinicopathological features and lactylation-related risk scores. In addition, the mRNA expression levels of five genes were verified in PCa cell lines by qPCR. Results: We identified 5 key LRGs (ALDOA, DDX39A, H2AX, KIF2C, RACGAP1) and constructed the LRGs prognostic model. The AUC values for 1 -, 3 -, and 5-year DFS in the TCGA dataset were 0.762, 0.745, and 0.709, respectively. The risk score was found a better predictor of DFS than traditional clinicopathological features in PCa. A nomogram that combined the risk score with clinical variables accurately predicted the outcome of the patients. The PCa patients in the high-risk group have a higher proportion of regulatory T cells and M2 macrophage, a higher tumor mutation burden, and a worse prognosis than those in the low-risk group. The high-risk group had a lower IC50 for certain chemotherapeutic drugs, such as Docetaxel, and Paclitaxel than the low-risk group. Furthermore, five key LRGs were found to be highly expressed in castration-resistant PCa cells. Conclusion: The lactylation-related genes prognostic model can effectively predict the DFS and therapeutic responses in patients with PCa.
Collapse
Affiliation(s)
| | | | | | | | - Zhigang Zhao
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, Guangzhou, China
| |
Collapse
|
3563
|
Flesken-Nikitin A, Pirtz MG, Ashe CS, Ellenson LH, Cosgrove BD, Nikitin AY. Dysregulation of cell state dynamics during early stages of serous endometrial carcinogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585274. [PMID: 38562813 PMCID: PMC10983873 DOI: 10.1101/2024.03.15.585274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Serous endometrial carcinoma (SEC) constitutes about 10% of endometrial carcinomas and is one of the most aggressive and lethal types of uterine cancer. Due to the rapid progression of SEC, early detection of this disease is of utmost importance. However, molecular and cellular dynamics during the pre-dysplastic stage of this disease remain largely unknown. Here, we provide a comprehensive census of cell types and their states for normal, pre-dysplastic, and dysplastic endometrium in a mouse model of SEC. This model is associated with inactivation of tumor suppressor genes Trp53 and Rb1 , whose pathways are altered frequently in SEC. We report that pre-dysplastic changes are characterized by an expanded and increasingly diverse immature luminal epithelial cell populations. Consistent with transcriptome changes, cells expressing the luminal epithelial marker TROP2 begin to substitute FOXA2+ cells in the glandular epithelium. These changes are associated with a reduction in number and strength of predicted interactions between epithelial and stromal endometrial cells. By using a multi-level approach combining single-cell and spatial transcriptomics paired with screening for clinically relevant genes in human endometrial carcinoma, we identified a panel of 44 genes suitable for further testing of their validity as early diagnostic and prognostic markers. Among these genes are known markers of human SEC, such as C DKN2A, and novel markers, such as OAS2 and OASL, members of 2-5A synthetase family that is essential for the innate immune response. In summary, our results suggest an important role of the luminal epithelium in SEC pathogenesis, highlight aberrant cell-cell interactions in pre-dysplastic stages, and provide a new platform for comparative identification and characterization of novel, clinically relevant prognostic and diagnostic markers and potential therapeutic modalities.
Collapse
|
3564
|
Chung DC, Gray DM, Singh H, Issaka RB, Raymond VM, Eagle C, Hu S, Chudova DI, Talasaz A, Greenson JK, Sinicrope FA, Gupta S, Grady WM. A Cell-free DNA Blood-Based Test for Colorectal Cancer Screening. N Engl J Med 2024; 390:973-983. [PMID: 38477985 DOI: 10.1056/nejmoa2304714] [Citation(s) in RCA: 90] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
BACKGROUND Colorectal cancer is the third most diagnosed cancer in adults in the United States. Early detection could prevent more than 90% of colorectal cancer-related deaths, yet more than one third of the screening-eligible population is not up to date with screening despite multiple available tests. A blood-based test has the potential to improve screening adherence, detect colorectal cancer earlier, and reduce colorectal cancer-related mortality. METHODS We assessed the performance characteristics of a cell-free DNA (cfDNA) blood-based test in a population eligible for colorectal cancer screening. The coprimary outcomes were sensitivity for colorectal cancer and specificity for advanced neoplasia (colorectal cancer or advanced precancerous lesions) relative to screening colonoscopy. The secondary outcome was sensitivity to detect advanced precancerous lesions. RESULTS The clinical validation cohort included 10,258 persons, 7861 of whom met eligibility criteria and were evaluable. A total of 83.1% of the participants with colorectal cancer detected by colonoscopy had a positive cfDNA test and 16.9% had a negative test, which indicates a sensitivity of the cfDNA test for detection of colorectal cancer of 83.1% (95% confidence interval [CI], 72.2 to 90.3). Sensitivity for stage I, II, or III colorectal cancer was 87.5% (95% CI, 75.3 to 94.1), and sensitivity for advanced precancerous lesions was 13.2% (95% CI, 11.3 to 15.3). A total of 89.6% of the participants without any advanced colorectal neoplasia (colorectal cancer or advanced precancerous lesions) identified on colonoscopy had a negative cfDNA blood-based test, whereas 10.4% had a positive cfDNA blood-based test, which indicates a specificity for any advanced neoplasia of 89.6% (95% CI, 88.8 to 90.3). Specificity for negative colonoscopy (no colorectal cancer, advanced precancerous lesions, or nonadvanced precancerous lesions) was 89.9% (95% CI, 89.0 to 90.7). CONCLUSIONS In an average-risk screening population, this cfDNA blood-based test had 83% sensitivity for colorectal cancer, 90% specificity for advanced neoplasia, and 13% sensitivity for advanced precancerous lesions. (Funded by Guardant Health; ECLIPSE ClinicalTrials.gov number, NCT04136002.).
Collapse
Affiliation(s)
- Daniel C Chung
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - Darrell M Gray
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - Harminder Singh
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - Rachel B Issaka
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - Victoria M Raymond
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - Craig Eagle
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - Sylvia Hu
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - Darya I Chudova
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - AmirAli Talasaz
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - Joel K Greenson
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - Frank A Sinicrope
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - Samir Gupta
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| | - William M Grady
- From the Division of Gastroenterology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston (D.C.C.); Gray Area Strategies, Owings Mills, MD (D.M.G.); the Association of Black Gastroenterologists and Hepatologists, New York (D.M.G.); the Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada (H.S.); the Divisions of Public Health Sciences (R.B.I., W.M.G.), Clinical Research (R.B.I.), and Translational Science and Therapeutics (W.M.G.), Fred Hutchinson Cancer Center, and the Division of Gastroenterology, University of Washington School of Medicine (R.B.I., W.M.G.) - both in Seattle; Guardant Health, Palo Alto (V.M.R., C.E., S.H., D.I.C., A.T.), and the University of California, San Diego, La Jolla (S.G.) - both in California; the Department of Pathology, Michigan Medicine, Ann Arbor (J.K.G.); and the Divisions of Oncology, Gastroenterology, and Hepatology, Mayo Clinic, Mayo Comprehensive Cancer Center and Mayo Alix School of Medicine, Rochester, MN (F.A.S.)
| |
Collapse
|
3565
|
Heard JR, Mitra AP. Noninvasive Tests for Bladder Cancer Detection and Surveillance: A Systematic Review of Commercially Available Assays. Bladder Cancer 2024; 10:71-81. [PMID: 38993533 PMCID: PMC11181788 DOI: 10.3233/blc-230096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/14/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND An important reason for the high health care costs associated with bladder cancer is the need for frequent cystoscopy for detection and surveillance of this disease. Cytologic analysis of voided urine specimens can assist, but is too inaccurate to replace cystoscopy. In an effort to create reliable, objective, noninvasive mechanisms for detecting bladder cancer, a number of urine-based molecular tests have been developed with the ultimate goal of reducing the frequency of cystoscopy. OBJECTIVE To summarize the performance of urine-based biomarker tests, currently commercially available in the US, as part of the initial workup for hematuria and for bladder cancer surveillance. METHODS In accordance with PRISMA guidelines we performed a systematic review of the literature on the performance of NMP22, BTA, UroVysion, ImmunoCyt/uCyt, CxBladder, and Bladder EpiCheck. Median sensitivity, specificity, negative (NPV) and positive predictive values (PPV) were calculated for each test based on the included studies. RESULTS Twenty-eight studies met inclusion criteria for the performance of five urine-based biomarker tests in the setting hematuria workup. Median sensitivity ranged from 65.7% -100% and specificity ranged from 62.5% -93.8%. Median NPV ranged from 94.2% -98.3% and PPV ranged from 29% -58.7%. Fourteen studies met inclusion criteria for the performance of six tests in the setting of bladder cancer surveillance. Median sensitivity ranged from 22.6% -92.0% and specificity from 20.5% -97.9%. Median NPV ranged from 52.9% -96.5% and PPV ranged from 48.1% -75.7%. CONCLUSIONS Our analysis finds that while these tests may provide some clinical utility, none of the assays have thus far demonstrated objective evidence to supplant the gold diagnostic standard.
Collapse
Affiliation(s)
- John R Heard
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anirban P Mitra
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
3566
|
Marino P, Mininni M, Deiana G, Marino G, Divella R, Bochicchio I, Giuliano A, Lapadula S, Lettini AR, Sanseverino F. Healthy Lifestyle and Cancer Risk: Modifiable Risk Factors to Prevent Cancer. Nutrients 2024; 16:800. [PMID: 38542712 PMCID: PMC10974142 DOI: 10.3390/nu16060800] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 08/07/2024] Open
Abstract
Cancer has become a serious problem worldwide, as it represents the main cause of death, and its incidence has increased over the years. A potential strategy to counter the growing spread of various forms of cancer is the adoption of prevention strategies, in particular, the use of healthy lifestyles, such as maintaining a healthy weight, following a healthy diet; being physically active; avoiding smoking, alcohol consumption, and sun exposure; and vitamin D supplementation. These modifiable risk factors are associated with this disease, contributing to its development, progression, and severity. This review evaluates the relationship between potentially modifiable risk factors and overall cancer development, specifically breast, colorectal, and prostate cancer, and highlights updated recommendations on cancer prevention. The results of numerous clinical and epidemiological studies clearly show the influence of lifestyles on the development and prevention of cancer. An incorrect diet, composed mainly of saturated fats and processed products, resulting in increased body weight, combined with physical inactivity, alcohol consumption, and smoking, has induced an increase in the incidence of all three types of cancer under study. Given the importance of adopting correct and healthy lifestyles to prevent cancer, global institutions should develop strategies and environments that encourage individuals to adopt healthy and regular behaviors.
Collapse
Affiliation(s)
- Pasquale Marino
- Unit of Oncological Gynecology, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Via Padre Pio, 1, 85028 Potenza, Italy; (G.D.); (F.S.)
| | - Mariangela Mininni
- Department Direzione Generale per la Salute e le Politiche della Persona, Regione Basilicata, Via Vincenzo Verrastro, 4, 85100 Potenza, Italy;
| | - Giovanni Deiana
- Unit of Oncological Gynecology, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Via Padre Pio, 1, 85028 Potenza, Italy; (G.D.); (F.S.)
| | - Graziella Marino
- Unit of Breast Surgery, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Potenza, Italy;
| | - Rosa Divella
- Nutritionist’s Studio at the Gravina in Puglia, C.so Giuseppe Di Vittorio, 14, 70024 Bari, Italy;
| | - Ilaria Bochicchio
- Unit of Clinical Psychology, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Via Padre Pio, 1, 85028 Potenza, Italy; (I.B.); (A.G.); (S.L.); (A.R.L.)
| | - Alda Giuliano
- Unit of Clinical Psychology, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Via Padre Pio, 1, 85028 Potenza, Italy; (I.B.); (A.G.); (S.L.); (A.R.L.)
| | - Stefania Lapadula
- Unit of Clinical Psychology, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Via Padre Pio, 1, 85028 Potenza, Italy; (I.B.); (A.G.); (S.L.); (A.R.L.)
| | - Alessandro Rocco Lettini
- Unit of Clinical Psychology, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Via Padre Pio, 1, 85028 Potenza, Italy; (I.B.); (A.G.); (S.L.); (A.R.L.)
| | - Francesca Sanseverino
- Unit of Oncological Gynecology, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Via Padre Pio, 1, 85028 Potenza, Italy; (G.D.); (F.S.)
| |
Collapse
|
3567
|
Larson AC, Knoche SM, Brumfield GL, Doty KR, Gephart BD, Moore-Saufley PR, Solheim JC. Gemcitabine Modulates HLA-I Regulation to Improve Tumor Antigen Presentation by Pancreatic Cancer Cells. Int J Mol Sci 2024; 25:3211. [PMID: 38542184 PMCID: PMC10970070 DOI: 10.3390/ijms25063211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/25/2024] [Accepted: 03/04/2024] [Indexed: 04/26/2024] Open
Abstract
Pancreatic cancer is a lethal disease, harboring a five-year overall survival rate of only 13%. Current treatment approaches thus require modulation, with attention shifting towards liberating the stalled efficacy of immunotherapies. Select chemotherapy drugs which possess inherent immune-modifying behaviors could revitalize immune activity against pancreatic tumors and potentiate immunotherapeutic success. In this study, we characterized the influence of gemcitabine, a chemotherapy drug approved for the treatment of pancreatic cancer, on tumor antigen presentation by human leukocyte antigen class I (HLA-I). Gemcitabine increased pancreatic cancer cells' HLA-I mRNA transcripts, total protein, surface expression, and surface stability. Temperature-dependent assay results indicated that the increased HLA-I stability may be due to reduced binding of low affinity peptides. Mass spectrometry analysis confirmed changes in the HLA-I-presented peptide pool post-treatment, and computational predictions suggested improved affinity and immunogenicity of peptides displayed solely by gemcitabine-treated cells. Most of the gemcitabine-exclusive peptides were derived from unique source proteins, with a notable overrepresentation of translation-related proteins. Gemcitabine also increased expression of select immunoproteasome subunits, providing a plausible mechanism for its modulation of the HLA-I-bound peptidome. Our work supports continued investigation of immunotherapies, including peptide-based vaccines, to be used with gemcitabine as new combination treatment modalities for pancreatic cancer.
Collapse
Affiliation(s)
- Alaina C. Larson
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shelby M. Knoche
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Gabrielle L. Brumfield
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kenadie R. Doty
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin D. Gephart
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | - Joyce C. Solheim
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
3568
|
Yang H, Li Y, Liang H, Cui C, Gan L, Zhang H. Design, Synthesis, Biological Evaluation and Molecular Docking of Novel F-18-Labeled Focal Adhesion Kinase Inhibitors as Potential Tumor Radiotracers. Molecules 2024; 29:1224. [PMID: 38542861 PMCID: PMC10974507 DOI: 10.3390/molecules29061224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 11/12/2024] Open
Abstract
Tumor diagnosis, especially at the early stages, holds immense significance. Focal adhesion kinase (FAK) is often highly expressed across various types of tumors, making it a promising target for both therapy and diagnosis. In this study, seven novel inhibitors were designed and synthesized. The inhibitory activity of these compounds against FAK was notably potent, with an IC50 range of 1.27-1968 nM. In particular, compounds 7a and 7c, with IC50 values of 5.59 nM and 1.27 nM, respectively, were radiolabeled with F-18 and then evaluated with S-180 tumor-bearing mice. Subsequently, they exhibited moderate-to-high tumor uptake values, with [18F]7a showing 1.39 ± 0.30%ID/g at 60 min post injection and [18F]7c demonstrating 6.58 ± 0.46%ID/g at 30 min post injection. In addition, the results from docking studies revealed the binding specifics of the studied compounds. Overall, these findings hold the potential to offer valuable guidance for enhancing the development of radiotracers and enzyme inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | - Huabei Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, College of Chemistry, Beijing Normal University, No. 19 Xinjiekouwai Street, Haidian District, Beijing 100875, China
| |
Collapse
|
3569
|
Bernal A, Bechler AJ, Mohan K, Rizzino A, Mathew G. The Current Therapeutic Landscape for Metastatic Prostate Cancer. Pharmaceuticals (Basel) 2024; 17:351. [PMID: 38543137 PMCID: PMC10974045 DOI: 10.3390/ph17030351] [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: 01/30/2024] [Revised: 02/16/2024] [Accepted: 03/05/2024] [Indexed: 04/01/2024] Open
Abstract
In 2024, there will be an estimated 1,466,718 cases of prostate cancer (PC) diagnosed globally, of which 299,010 cases are estimated to be from the US. The typical clinical approach for PC involves routine screening, diagnosis, and standard lines of treatment. However, not all patients respond to therapy and are subsequently diagnosed with treatment emergent neuroendocrine prostate cancer (NEPC). There are currently no approved treatments for this form of aggressive PC. In this review, a compilation of the clinical trials regimen to treat late-stage NEPC using novel targets and/or a combination approach is presented. The novel targets assessed include DLL3, EZH2, B7-H3, Aurora-kinase-A (AURKA), receptor tyrosine kinases, PD-L1, and PD-1. Among these, the trials administering drugs Alisertib or Cabozantinib, which target AURKA or receptor tyrosine kinases, respectively, appear to have promising results. The least effective trials appear to be ones that target the immune checkpoint pathways PD-1/PD-L1. Many promising clinical trials are currently in progress. Consequently, the landscape of successful treatment regimens for NEPC is extremely limited. These trial results and the literature on the topic emphasize the need for new preventative measures, diagnostics, disease specific biomarkers, and a thorough clinical understanding of NEPC.
Collapse
Affiliation(s)
- Anastasia Bernal
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68106, USA; (A.B.); (A.J.B.); (K.M.); (A.R.)
| | - Alivia Jane Bechler
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68106, USA; (A.B.); (A.J.B.); (K.M.); (A.R.)
| | - Kabhilan Mohan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68106, USA; (A.B.); (A.J.B.); (K.M.); (A.R.)
| | - Angie Rizzino
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68106, USA; (A.B.); (A.J.B.); (K.M.); (A.R.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68106, USA
| | - Grinu Mathew
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68106, USA; (A.B.); (A.J.B.); (K.M.); (A.R.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68106, USA
| |
Collapse
|
3570
|
Hughes MS, Addala A, Buckingham B. Digital Technology for Diabetes. Reply. N Engl J Med 2024; 390:963-964. [PMID: 38446694 DOI: 10.1056/nejmc2315000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
|
3571
|
Chiappa M, Guffanti F, Grasselli C, Panini N, Corbelli A, Fiordaliso F, Damia G. Different Patterns of Platinum Resistance in Ovarian Cancer Cells with Homologous Recombination Proficient and Deficient Background. Int J Mol Sci 2024; 25:3049. [PMID: 38474294 DOI: 10.3390/ijms25053049] [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: 01/19/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Platinum compounds are very active in first-line treatments of ovarian carcinoma. In fact, high rates of complete remission are achieved, but most patients eventually relapse with resistant disease. Many mechanisms underlying the platinum-resistant phenotype have been reported. However, there are no data in the same isogenic cell system proficient and deficient in homologous recombination (HR) on platinum-acquired resistance that might unequivocally clarify the most important mechanism associated with resistance. We generated and characterized cisplatin (DDP)-resistant murine ovarian ID8 cell lines in a HR-deficient and -proficient background. Specific upregulation of the NER pathway in the HR-proficient and -resistant cells and partial restoration of HR in Brca1-/--resistant cells were found. Combinations of different inhibitors of the DNA damage response pathways with cisplatin were strongly active in both resistant and parental cells. The data from the ID8 isogenic system are in line with current experimental and clinical evidence and strongly suggest that platinum resistance develops in different ways depending on the cell DNA repair status (i.e., HR-proficient or HR-deficient), and the upregulation and/or restoration of repair pathways are major determinants of DDP resistance.
Collapse
Affiliation(s)
- Michela Chiappa
- Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Federica Guffanti
- Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Chiara Grasselli
- Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Nicolò Panini
- Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Alessandro Corbelli
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Fabio Fiordaliso
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Giovanna Damia
- Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| |
Collapse
|
3572
|
Beatrici E, Frego N, Chiarelli G, Sordelli F, Mancon S, Saitta C, De Carne F, Garofano G, Arena P, Avolio PP, Gobbo A, Uleri A, Contieri R, Paciotti M, Lazzeri M, Hurle R, Casale P, Buffi NM, Lughezzani G. A Comparative Evaluation of Multiparametric Magnetic Resonance Imaging and Micro-Ultrasound for the Detection of Clinically Significant Prostate Cancer in Patients with Prior Negative Biopsies. Diagnostics (Basel) 2024; 14:525. [PMID: 38472997 DOI: 10.3390/diagnostics14050525] [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: 01/30/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND The diagnostic process for prostate cancer after a negative biopsy is challenging. This study compares the diagnostic accuracy of micro-ultrasound (mUS) with multiparametric magnetic resonance imaging (mpMRI) for such cases. METHODS A retrospective cohort study was performed, targeting men with previous negative biopsies and using mUS and mpMRI to detect prostate cancer and clinically significant prostate cancer (csPCa). RESULTS In our cohort of 1397 men, 304 had a history of negative biopsies. mUS was more sensitive than mpMRI, with better predictive value for negative results. Importantly, mUS was significantly associated with csPCa detection (adjusted odds ratio [aOR]: 6.58; 95% confidence interval [CI]: 1.15-37.8; p = 0.035). CONCLUSIONS mUS may be preferable for diagnosing prostate cancer in previously biopsy-negative patients. However, the retrospective design of this study at a single institution suggests that further research across multiple centers is warranted.
Collapse
Affiliation(s)
- Edoardo Beatrici
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Nicola Frego
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Giuseppe Chiarelli
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Federica Sordelli
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Stefano Mancon
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Cesare Saitta
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Fabio De Carne
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Giuseppe Garofano
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Paola Arena
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Pier Paolo Avolio
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Andrea Gobbo
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Alessandro Uleri
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Roberto Contieri
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Marco Paciotti
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Massimo Lazzeri
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Rodolfo Hurle
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Paolo Casale
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Nicolò Maria Buffi
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Giovanni Lughezzani
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Department of Urology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| |
Collapse
|
3573
|
Lin Q, Serratore A, Niu J, Shen S, Roy Chaudhuri T, Ma WW, Qu J, Kandel ES, Straubinger RM. Fibroblast growth factor receptor 1 inhibition suppresses pancreatic cancer chemoresistance and chemotherapy-driven aggressiveness. Drug Resist Updat 2024; 73:101064. [PMID: 38387284 PMCID: PMC11864563 DOI: 10.1016/j.drup.2024.101064] [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: 09/30/2023] [Revised: 12/26/2023] [Accepted: 02/01/2024] [Indexed: 02/24/2024]
Abstract
AIMS Pancreatic ductal adenocarcinoma (PDAC) is often intrinsically-resistant to standard-of-care chemotherapies such as gemcitabine. Acquired gemcitabine resistance (GemR) can arise from treatment of initially-sensitive tumors, and chemotherapy can increase tumor aggressiveness. We investigated the molecular mechanisms of chemoresistance and chemotherapy-driven tumor aggressiveness, which are understood incompletely. METHODS Differential proteomic analysis was employed to investigate chemotherapy-driven chemoresistance drivers and responses of PDAC cells and patient-derived tumor xenografts (PDX) having different chemosensitivities. We also investigated the prognostic value of FGFR1 expression in the efficacy of selective pan-FGFR inhibitor (FGFRi)-gemcitabine combinations. RESULTS Quantitative proteomic analysis of a highly-GemR cell line revealed fibroblast growth factor receptor 1 (FGFR1) as the highest-expressed receptor tyrosine kinase. FGFR1 knockdown or FGFRi co-treatment enhanced gemcitabine efficacy and decreased GemR marker expression, implicating FGFR1 in augmentation of GemR. FGFRi treatment reduced PDX tumor progression and prolonged survival significantly, even in highly-resistant tumors in which neither single-agent showed efficacy. Gemcitabine exacerbated aggressiveness of highly-GemR tumors, based upon proliferation and metastatic markers. Combining FGFRi with gemcitabine or gemcitabine+nab-paclitaxel reversed tumor aggressiveness and progression, and prolonged survival significantly. In multiple PDAC PDXs, FGFR1 expression correlated with intrinsic tumor gemcitabine sensitivity. CONCLUSION FGFR1 drives chemoresistance and tumor aggressiveness, which FGFRi can reverse.
Collapse
Affiliation(s)
- Qingxiang Lin
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; New York State Center of Excellence in Bioinformatics & Life Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Andrea Serratore
- New York State Center of Excellence in Bioinformatics & Life Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Jin Niu
- New York State Center of Excellence in Bioinformatics & Life Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Shichen Shen
- New York State Center of Excellence in Bioinformatics & Life Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Tista Roy Chaudhuri
- New York State Center of Excellence in Bioinformatics & Life Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Wen Wee Ma
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jun Qu
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; New York State Center of Excellence in Bioinformatics & Life Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Eugene S Kandel
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Robert M Straubinger
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; New York State Center of Excellence in Bioinformatics & Life Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA; Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
3574
|
Eddy K, Gupta K, Eddin MN, Marinaro C, Putta S, Sauer JM, Chaly A, Freeman KB, Pelletier JC, Fateeva A, Furmanski P, Silk AW, Reitz AB, Zloza A, Chen S. Assessing Longitudinal Treatment Efficacies and Alterations in Molecular Markers Associated with Glutamatergic Signaling and Immune Checkpoint Inhibitors in a Spontaneous Melanoma Mouse Model. JID INNOVATIONS 2024; 4:100262. [PMID: 38445232 PMCID: PMC10914525 DOI: 10.1016/j.xjidi.2024.100262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 03/07/2024] Open
Abstract
Previous work done by our laboratory described the use of an immunocompetent spontaneous melanoma-prone mouse model, TGS (TG-3/SKH-1), to evaluate treatment outcomes using inhibitors of glutamatergic signaling and immune checkpoint for 18 weeks. We showed a significant therapeutic efficacy with a notable sex-biased response in male mice. In this follow-up 18-week study, the dose of the glutamatergic signaling inhibitor was increased (from 1.7 mg/kg to 25 mg/kg), which resulted in improved responses in female mice but not male mice. The greatest reduction in tumor progression was observed in male mice treated with single-agent troriluzole and anti-PD-1. Furthermore, a randomly selected group of mice was removed from treatment after 18 weeks and maintained for up to an additional 48 weeks demonstrating the utility of the TGS mouse model to perform a ≥1-year preclinical therapeutic study in a physiologically relevant tumor-host environment. Digital spatial imaging analyses were performed in tumors and tumor microenvironments across treatment modalities using antibody panels for immune cell types and immune cell activation. The results suggest that immune cell populations and cytotoxic activities of T cells play critical roles in treatment responses in these mice. Examination of a group of molecular protein markers based on the proposed mechanisms of action of inhibitors of glutamatergic signaling and immune checkpoint showed that alterations in expression levels of xCT, γ-H2AX, EAAT2, PD-L1, and PD-1 are likely associated with the loss of treatment responses. These results suggest the importance of tracking changes in molecular markers associated with the mechanism of action of therapeutics over the course of a longitudinal preclinical therapeutic study in spatial and temporal manners.
Collapse
Affiliation(s)
- Kevinn Eddy
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
- Graduate Program in Cellular & Molecular Pharmacology, School of Graduate Studies, Rutgers University, Piscataway, New Jersey, USA
| | - Kajal Gupta
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois, USA
| | - Mohamad Naser Eddin
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Christina Marinaro
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Sanjana Putta
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - John Michael Sauer
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Anna Chaly
- Fox Chase Therapeutics Discovery, Doylestown, Pennsylvania, USA
| | | | | | - Anna Fateeva
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
- Graduate Program in Cellular & Molecular Pharmacology, School of Graduate Studies, Rutgers University, Piscataway, New Jersey, USA
| | - Philip Furmanski
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
- Environmental & Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Ann W. Silk
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Allen B. Reitz
- Fox Chase Therapeutics Discovery, Doylestown, Pennsylvania, USA
| | - Andrew Zloza
- Division of Hematology, Oncology, and Cell Therapy, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Suzie Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
- Graduate Program in Cellular & Molecular Pharmacology, School of Graduate Studies, Rutgers University, Piscataway, New Jersey, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
- Environmental & Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
3575
|
Ebaid NF, Abdelkawy KS, Shehata MA, Salem HF, Magdy G, Hussein RRS, Elbarbry F. Effects of pharmacogenetics on pharmacokinetics and toxicity of doxorubicin in Egyptian breast cancer patients. Xenobiotica 2024; 54:160-170. [PMID: 38491961 DOI: 10.1080/00498254.2024.2330493] [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: 01/30/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
This study investigates the impact of single nucleotide polymorphisms in genes (SLC22A16 and CBR1) involved in the pharmacokinetics and toxicity of doxorubicin (DOX) in Egyptian female patients with breast cancer.Patients administered DOX (60 mg/m2) for 4 cycles every 3 weeks. The peak DOX plasma concentration was measured using a validated chromatographic method. The genotyping for the selected SNPs, SLC22A16 T > C (rs714368), and CBR1 C > T (rs20572), was performed by RT-PCR. Patients were monitored for hematological and cardiac toxicities.The variant carriers of CBR1 C > T (rs20572) exhibited significantly higher DOX concentration, but no significant association to DOX-induced hematological toxicity. On the other hand, SLC22A16 T > C (rs714368) had no significant influence on DOX plasma concentration, but was significantly correlated with lower risk of neutropenia (OR 0.31, 95% CI 0.12-0.75, p = 0.01) and leukopoenia (OR 0.18, 95% CI 0.07-0.5, p = 0.001). DOX-related cardiotoxicity was correlated with the cumulative dose of DOX (R = 0.238, p = 0.017), but not with any of the two examined SNPs.Genetic polymorphisms in SLC22A16 and CBR1 may explain the inter-individual variations in DOX pharmacokinetics and toxicity. Using pharmacogenetic testing is important to customise drug therapy for cancer patients treated with anthracyclines.
Collapse
Affiliation(s)
- N F Ebaid
- Clinical Pharmacy Department, Faculty of Pharmacy, Menoufia University, Al Minufiyah, Egypt
| | - K S Abdelkawy
- Clinical Pharmacy Department, Faculty of Pharmacy, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - M A Shehata
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Menoufia University, Al Minufiyah, Egypt
| | - H F Salem
- Pharmaceutics and Industrial Pharmacy Department, Beni-Suef University, Beni Suef, Egypt
| | - G Magdy
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - R R S Hussein
- Clinical Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
| | - F Elbarbry
- Pacific University School of Pharmacy, Hillsboro, OR, USA
| |
Collapse
|
3576
|
Ledford H. Why are so many young people getting cancer? What the data say. Nature 2024; 627:258-260. [PMID: 38480960 DOI: 10.1038/d41586-024-00720-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
|
3577
|
Jiang Y, Abboud Y, Liang J, Larson B, Osipov A, Gong J, Hendifar AE, Atkins K, Liu Q, Nissen NN, Li D, Pandol SJ, Lo SK, Gaddam S. The Disproportionate Rise in Pancreatic Cancer in Younger Women Is Due to a Rise in Adenocarcinoma and Not Neuroendocrine Tumors: A Nationwide Time-Trend Analysis Using 2001-2018 United States Cancer Statistics Databases. Cancers (Basel) 2024; 16:971. [PMID: 38473332 DOI: 10.3390/cancers16050971] [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: 01/28/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
In previous studies, a significant increase in the incidence of pancreatic cancer among younger women compared to men in the United States was noted. However, the specific histopathologic characteristics were not delineated. This population-based study aimed to assess whether this disproportionate rise in pancreatic cancer in younger women was contributed by pancreatic ductal adenocarcinoma (PDAC) or pancreatic neuroendocrine tumors (PanNET). The United States Cancer Statistics (USCS) database was used to identify patients with pancreatic cancer between 2001 and 2018. The results showed that, in younger adults, the incidence of PDAC has increased in women [average annual percentage change (AAPC) = 0.62%], while it has remained stable in men (AAPC = -0.09%). The PDAC incidence rate among women increased at a greater rate compared to men with a statistically significant difference in AAPC (p < 0.001), with neither identical nor parallel trends. In contrast, cases of PanNET did not demonstrate a statistically significant sex-specific AAPC difference. In conclusion, this study demonstrated that the dramatic increase in the incidence rate of PDAC explains the disproportionate rise in pancreatic cancer incidence in younger women. This prompts further prospective studies to investigate the underlying reasons for these sex-specific disparities in PDAC.
Collapse
Affiliation(s)
- Yi Jiang
- Karsh Division of Gastroenterology, Departments of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yazan Abboud
- Karsh Division of Gastroenterology, Departments of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jeff Liang
- Karsh Division of Gastroenterology, Departments of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Brent Larson
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Arsen Osipov
- Division of Medical Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jun Gong
- Division of Medical Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrew E Hendifar
- Division of Medical Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Katelyn Atkins
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Quin Liu
- Karsh Division of Gastroenterology, Departments of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nicholas N Nissen
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Debiao Li
- Biomedical Imaging Research Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Stephen J Pandol
- Karsh Division of Gastroenterology, Departments of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Simon K Lo
- Karsh Division of Gastroenterology, Departments of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Srinivas Gaddam
- Karsh Division of Gastroenterology, Departments of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
3578
|
Montoro-Jiménez I, Granda-Díaz R, Menéndez ST, Prieto-Fernández L, Otero-Rosales M, Álvarez-González M, García-de-la-Fuente V, Rodríguez A, Rodrigo JP, Álvarez-Teijeiro S, García-Pedrero JM, Hermida-Prado F. Combined PIK3CA and SOX2 Gene Amplification Predicts Laryngeal Cancer Risk beyond Histopathological Grading. Int J Mol Sci 2024; 25:2695. [PMID: 38473941 DOI: 10.3390/ijms25052695] [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: 12/31/2023] [Revised: 02/05/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
The PIK3CA and SOX2 genes map at 3q26, a chromosomal region frequently amplified in head and neck cancers, which is associated with poor prognosis. This study explores the clinical significance of PIK3CA and SOX2 gene amplification in early tumorigenesis. Gene copy number was analyzed by real-time PCR in 62 laryngeal precancerous lesions and correlated with histopathological grading and laryngeal cancer risk. Amplification of the SOX2 and PIK3CA genes was frequently detected in 19 (31%) and 32 (52%) laryngeal dysplasias, respectively, and co-amplification in 18 (29%) cases. The PIK3CA and SOX2 amplifications were predominant in high-grade dysplasias and significantly associated with laryngeal cancer risk beyond histological criteria. Multivariable Cox analysis further revealed PIK3CA gene amplification as an independent predictor of laryngeal cancer development. Interestingly, combined PIK3CA and SOX2 amplification allowed us to distinguish three cancer risk subgroups, and PIK3CA and SOX2 co-amplification was found the strongest predictor by ROC analysis. Our data demonstrate the clinical relevance of PIK3CA and SOX2 amplification in early laryngeal tumorigenesis. Remarkably, PIK3CA amplification was found to be an independent cancer predictor. Furthermore, combined PIK3CA and SOX2 amplification is emerging as a valuable and easy-to-implement tool for cancer risk assessment in patients with laryngeal precancerous lesions beyond current WHO histological grading.
Collapse
Affiliation(s)
- Irene Montoro-Jiménez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rocío Granda-Díaz
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sofía T Menéndez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Llara Prieto-Fernández
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Otero-Rosales
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel Álvarez-González
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
| | - Vanessa García-de-la-Fuente
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
| | - Aida Rodríguez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
| | - Juan P Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Saúl Álvarez-Teijeiro
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juana M García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco Hermida-Prado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33011 Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
3579
|
Piotrowska A, Nowak JI, Wierzbicka JM, Domżalski P, Górska-Arcisz M, Sądej R, Popiel D, Wieczorek M, Żmijewski MA. Fibroblast Growth Factor Receptor Inhibitors Decrease Proliferation of Melanoma Cell Lines and Their Activity Is Modulated by Vitamin D. Int J Mol Sci 2024; 25:2505. [PMID: 38473753 DOI: 10.3390/ijms25052505] [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: 01/25/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/14/2024] Open
Abstract
Regardless of the unprecedented progress in malignant melanoma treatment strategies and clinical outcomes of patients during the last twelve years, this skin cancer remains the most lethal one. We have previously documented that vitamin D and its low-calcaemic analogues enhance the anticancer activity of drugs including a classic chemotherapeutic-dacarbazine-and an antiangiogenic VEGFRs inhibitor-cediranib. In this study, we explored the response of A375 and RPMI7951 melanoma lines to CPL304110 (CPL110), a novel selective inhibitor of fibroblast growth factor receptors (FGFRs), and compared its efficacy with that of AZD4547, the first-generation FGFRs selective inhibitor. We also tested whether 1,25(OH)2D3, the active form of vitamin D, modulates the response of the cells to these drugs. CPL304110 efficiently decreased the viability of melanoma cells in both A375 and RPMI7951 cell lines, with the IC50 value below 1 µM. However, the metastatic RPMI7951 melanoma cells were less sensitive to the tested drug than A375 cells, isolated from primary tumour site. Both tested FGFR inhibitors triggered G0/G1 cell cycle arrest in A375 melanoma cells and increased apoptotic/necrotic SubG1 fraction in RPMI7951 melanoma cells. 1,25(OH)2D3 modulated the efficacy of CPL304110, by decreasing the IC50 value by more than 4-fold in A375 cell line, but not in RPMI7951 cells. Further analysis revealed that both inhibitors impact vitamin D signalling to some extent, and this effect is cell line-specific. On the other hand, 1,25(OH)2D3, have an impact on the expression of FGFR receptors and phosphorylation (FGFR-Tyr653/654). Interestingly, 1,25(OH)2D3 and CPL304110 co-treatment resulted in activation of the ERK1/2 pathway in A375 cells. Our results strongly suggested possible crosstalk between vitamin D-activated pathways and activity of FGFR inhibitors, which should be considered in further clinical studies.
Collapse
Affiliation(s)
- Anna Piotrowska
- Faculty of Medicine, Department of Histology, Medical University of Gdańsk, Dębinki 1a, 80-384 Gdańsk, Poland
| | - Joanna I Nowak
- Faculty of Medicine, Department of Histology, Medical University of Gdańsk, Dębinki 1a, 80-384 Gdańsk, Poland
| | - Justyna M Wierzbicka
- Faculty of Medicine, Department of Histology, Medical University of Gdańsk, Dębinki 1a, 80-384 Gdańsk, Poland
| | - Paweł Domżalski
- Faculty of Medicine, Department of Histology, Medical University of Gdańsk, Dębinki 1a, 80-384 Gdańsk, Poland
| | - Monika Górska-Arcisz
- Laboratory of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1, 80-384 Gdańsk, Poland
| | - Rafał Sądej
- Laboratory of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1, 80-384 Gdańsk, Poland
| | - Delfina Popiel
- Preclinical Development Departament, Celon Pharma S.A., Marymoncka 15, 05-152 Kazuń Nowy, Poland
| | - Maciej Wieczorek
- Preclinical Development Departament, Celon Pharma S.A., Marymoncka 15, 05-152 Kazuń Nowy, Poland
- Clinical Development Department, Celon Pharma S.A., Marymoncka 15, 05-152 Kazuń Nowy, Poland
| | - Michał A Żmijewski
- Faculty of Medicine, Department of Histology, Medical University of Gdańsk, Dębinki 1a, 80-384 Gdańsk, Poland
| |
Collapse
|
3580
|
Araya M, Sepúlveda F, Villegas J, Alarcón L, Burzio LO, Burzio VA, Borgna V. Knockdown of Antisense Noncoding Mitochondrial RNA Reduces Tumorigenicity of Patient-Derived Clear Cell Renal Carcinoma Cells in an Orthotopic Xenograft Mouse Model. Cancers (Basel) 2024; 16:830. [PMID: 38398221 PMCID: PMC10886546 DOI: 10.3390/cancers16040830] [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: 12/31/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent form of renal cancer and its treatment is hindered by a resistance to targeted therapies, immunotherapies and combinations of both. We have reported that the knockdown of the antisense noncoding mitochondrial RNAs (ASncmtRNAs) with chemically modified antisense oligonucleotides induces proliferative arrest and apoptotic death in tumor cells from many human and mouse cancer types. These studies have been mostly performed in vitro and in vivo on commercially available cancer cell lines and have shown that in mouse models tumor growth is stunted by the treatment. The present work was performed on cells derived from primary and metastatic ccRCC tumors. We established primary cultures from primary and metastatic ccRCC tumors, which were subjected to knockdown of ASncmtRNAs in vitro and in vivo in an orthotopic xenograft model in NOD/SCID mice. We found that these primary ccRCC cells are affected in the same way as tumor cell lines and in the orthotopic model tumor growth was significantly reduced by the treatment. This study on patient-derived ccRCC tumor cells represents a model closer to actual patient ccRCC tumors and shows that knockdown of ASncmtRNAs poses a potential treatment option for these patients.
Collapse
Affiliation(s)
- Mariela Araya
- Centro Cientifico & Tecnologico de Excelencia Ciencia & Vida, Santiago 8580702, Chile;
| | - Francisca Sepúlveda
- Center for Regenerative Medicine, Faculty of Clinical Medicine, Clínica Alemana, Universidad del Desarrollo, Santiago 7610615, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380492, Chile
| | - Jaime Villegas
- School of Veterinary Medicine, Faculty of Life Sciences, Universidad Andrés Bello, Santiago 8370251, Chile;
| | - Luis Alarcón
- Urology Service, Hospital Barros Luco-Trudeau, Santiago 8900085, Chile;
| | - Luis O. Burzio
- Department of Biological Sciences, Faculty of Life Sciences, Universidad Andrés Bello, Santiago 8370251, Chile;
| | - Verónica A. Burzio
- Department of Biological Sciences, Faculty of Life Sciences, Universidad Andrés Bello, Santiago 8370251, Chile;
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago 8370134, Chile
| | - Vincenzo Borgna
- Centro Cientifico & Tecnologico de Excelencia Ciencia & Vida, Santiago 8580702, Chile;
- Urology Service, Hospital Barros Luco-Trudeau, Santiago 8900085, Chile;
- School of Medicine, Faculty of Medical Sciences, Universidad de Santiago de Chile, Santiago 9170022, Chile
- Faculty of Medicine and Science, Universidad San Sebastián, Santiago 7510602, Chile
| |
Collapse
|
3581
|
Zimmerman BS, Esteva FJ. Next-Generation HER2-Targeted Antibody-Drug Conjugates in Breast Cancer. Cancers (Basel) 2024; 16:800. [PMID: 38398191 PMCID: PMC10887217 DOI: 10.3390/cancers16040800] [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: 01/07/2024] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) tyrosine kinase is overexpressed in 20% of breast cancers and associated with a less favorable prognosis compared to HER2-negative disease. Patients have traditionally been treated with a combination of chemotherapy and HER2-targeted monoclonal antibodies such as trastuzumab and pertuzumab. The HER2-targeted antibody-drug conjugates (ADCs) trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd) represent a novel class of therapeutics in breast cancer. These drugs augment monoclonal antibodies with a cytotoxic payload, which is attached by a linker, forming the basic structure of an ADC. Novel combinations and sequential approaches are under investigation to overcome resistance to T-DM1 and T-DXd. Furthermore, the landscape of HER2-targeted therapy is rapidly advancing with the development of ADCs designed to attack cancer cells with greater precision and reduced toxicity. This review provides an updated summary of the current state of HER2-targeted ADCs as well as a detailed review of investigational agents on the horizon. Clinical trials are crucial in determining the optimal dosing regimens, understanding resistance mechanisms, and identifying patient populations that would derive the most benefit from these treatments. These novel ADCs are at the forefront of a new era in targeted cancer therapy, holding the potential to improve outcomes for patients with HER2-positive and HER2-Low breast cancer.
Collapse
Affiliation(s)
- Brittney S. Zimmerman
- Northwell, New Hyde Park, NY 11042, USA;
- Northwell Health Cancer Institute, Lake Success, NY 11042, USA
| | - Francisco J. Esteva
- Northwell, New Hyde Park, NY 11042, USA;
- Northwell Health Cancer Institute, Lake Success, NY 11042, USA
| |
Collapse
|
3582
|
Abstract
Bladder cancer remains a leading cause of cancer death worldwide and is associated with substantial impacts on patient quality of life, morbidity, mortality, and cost to the healthcare system. Gross hematuria frequently precedes the diagnosis of bladder cancer. Non-muscle-invasive bladder cancer (NMIBC) is managed initially with transurethral resection of a bladder tumor (TURBT), followed by a risk stratified approach to adjuvant intravesical therapy (IVe), and is associated with an overall survival of 90%. However, cure rates remain lower for muscle invasive bladder cancer (MIBC) owing to a variety of factors. NMIBC and MIBC groupings are heterogeneous and have unique pathological and molecular characteristics. Indeed, The Cancer Genome Atlas project identified genetic drivers and luminal and basal molecular subtypes of MIBC with distinct treatment responses. For NMIBC, IVe immunotherapy (primarily BCG) is the gold standard treatment for high grade and high risk NMIBC to reduce or prevent both recurrence and progression after initial TURBT; novel trials incorporate immune checkpoint inhibitors. IVe gene therapy and combination IVe chemotherapy have recently been completed, with promising results. For localized MIBC, essential goals are improving care and reducing morbidity following cystectomy or bladder preserving strategies. In metastatic disease, advances in understanding of the genomic landscape and tumor microenvironment have led to the implementation of immune checkpoint inhibitors, targeted treatments, and antibody-drug conjugates. Defining better selection criteria to identify the patients most likely to benefit from a specific treatment is an urgent need.
Collapse
Affiliation(s)
- Antonio Lopez-Beltran
- Department of Morphological Sciences, Unit of Anatomic Pathology, University of Cordoba Medical School, Cordoba, Spain
| | - Michael S Cookson
- Department of Urology, University of Oklahoma Health Sciences Center and the Stephenson Cancer Center, Oklahoma City, OK, US
| | - Brendan J Guercio
- Department of Medicine, James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, US
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Department of Surgery (Urology), Brown University, Providence, RI, US
- Legorreta Cancer Center, Brown University
- Lifespan Health Care System, Brown University
| |
Collapse
|
3583
|
Francescone R, Crawford HC, Vendramini-Costa DB. Rethinking the Roles of Cancer-Associated Fibroblasts in Pancreatic Cancer. Cell Mol Gastroenterol Hepatol 2024; 17:737-743. [PMID: 38316215 PMCID: PMC10966284 DOI: 10.1016/j.jcmgh.2024.01.022] [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: 11/19/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Bearing a dismal 5-year survival rate, pancreatic ductal adenocarcinoma (PDAC) is a challenging disease that features a unique fibroinflammatory tumor microenvironment. As major components of the PDAC tumor microenvironment, cancer-associated fibroblasts are still poorly understood and their contribution to the several hallmarks of PDAC, such as resistance to therapies, immunosuppression, and high incidence of metastasis, is likely underestimated. There have been encouraging advances in the understanding of these fascinating cells, but many controversies remain, leaving the field still actively exploring the full scope of their contributions in PDAC progression. Here we pose several important considerations regarding PDAC cancer-associated fibroblast functions. We posit that transcriptomic analyses be interpreted with caution, when aiming to uncover the functional contributions of these cells. Moreover, we propose that normalizing these functions, rather than eliminating them, will provide the opportunity to enhance therapeutic response. Finally, we propose that cancer-associated fibroblasts should not be studied in isolation, but in conjunction with its extracellular matrix, because their respective functions are coordinated and concordant.
Collapse
Affiliation(s)
- Ralph Francescone
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Howard C Crawford
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Debora Barbosa Vendramini-Costa
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan.
| |
Collapse
|
3584
|
Pankratz VS, Kanda D, Kosich M, Edwardson N, English K, Adsul P, Mishra SI. Racial and Ethnic Disparities in Colorectal Cancer Incidence Trends Across Regions of the United States From 2001 to 2020 - A United States Cancer Statistics Analysis. Cancer Control 2024; 31:10732748241300653. [PMID: 39543981 PMCID: PMC11565617 DOI: 10.1177/10732748241300653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/20/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) incidence rates have been decreasing in the United States (US), but there is limited information about differences in these improvements among individuals from different racial and ethnic subgroups across different regions of the US. METHODS Data from the National Program of Cancer Registries (NPCR) and the Surveillance, Epidemiology, and End Results (SEER) databases were used to examine trends in CRC incidence from 2001 to 2020 using a population-based retrospective cohort study. We obtained annual estimates of CRC incidence and used meta-regression analyses via weighted linear models to identify main effects and interactions that explained differences in CRC incidence trends among groups defined by race/ethnicity and US region while also considering CRC stage and sex. To summarize overall trends over time in incidence rates for specific racial and ethnic groups within and across US regions, we obtained average annual percentage change (AAPC) estimates. RESULTS The greatest differences in CRC incidence trends were among groups defined by race/ethnicity and US region. Non-Hispanic Black (NHB) persons had the largest declines in CRC incidence, with AAPC estimates ranging from -2.27 (95% CI: -2.49 to -2.06) in the South to -3.03 (95% CI: -3.59 to -2.47) in the West, but had higher-than-average incidence rates at study end. The AAPC estimate for American Indian/Alaska Native (AIAN) persons suggested no significant change over time (AAPC: -0.41, 95% CI: -2.51 to 1.73). CONCLUSION CRC incidence trends differ among racial/ethnic groups residing in different US regions. Notably, CRC incidence rates have not changed noticeably for AIAN persons from 2001-2020. These findings highlight the importance of reinvigorating collaborative efforts to develop geographic and population-specific screening and preventative approaches to reduce the CRC burden experienced by Native American communities and members of other minoritized groups.
Collapse
Affiliation(s)
- Vernon Shane Pankratz
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Deborah Kanda
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Mikaela Kosich
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Nicholas Edwardson
- School of Public Administration, University of New Mexico, Albuquerque, NM, USA
| | - Kevin English
- Albuquerque Area Southwest Tribal Epidemiology Center, Albuquerque Area Indian Health Board, Inc., Albuquerque, NM, USA
| | - Prajakta Adsul
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Shiraz I. Mishra
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Family and Community Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
3585
|
Ehman KM, Jenkins GD, Grimm JA, Cammack TL, Samuelson BJ, Stoll RJ, Maxson JA, MacLaughlin KL. Primary Human Papillomavirus Test Uptake and Cervical Cancer Screening Trends in the Midwest, United States. J Prim Care Community Health 2024; 15:21501319241251934. [PMID: 38726652 PMCID: PMC11085000 DOI: 10.1177/21501319241251934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/21/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
INTRODUCTION/OBJECTIVES Despite U.S. Preventive Services Task Force and American Cancer Society endorsement of primary HPV screening, limited published data shows low uptake. PRIMARY AIM Assess cervical cancer screening rates over time, particularly primary HPV test uptake, among patients in a midwestern practice. SECONDARY AIM Evaluate associations between sociodemographics and screening adherence. METHODS Cross-sectional study. Qualifying subjects and type of screening test used were identified by applying ICD-9, ICD-10, lab test, and CPT codes to the Unified Data Platform. Sociodemographics were found through the electronic health record. RESULTS Primary HPV uptake represented <1% of annual screening from 1/2017 to 1/2022. On 1/1/2022, only 55% of 21 to 29 year old and 63% of 30 to 65 year old were up to date with screening among the studied population. For 21 to 29 year old, compared with White women, Black women were 28% less likely to be screened [RR = 0.72 (0.66-0.79)]. Compared with never-smokers, current smokers were 9% less likely to be screened [RR = 0.91 (0.87-0.96)], past smokers were 14% more likely [RR = 1.14 (1.09-1.2)]. Among 30 to 65 year old, compared with White women, Black women were 14% less likely to be screened [RR = 0.86 (0.81-0.9)]. Compared with never-smokers, current smokers were 21% less likely to be screened [RR = 0.79 (0.77-0.81)], past smokers were 6% less likely [RR = 0.94 (0.92-0.95)]. Jointly considering race, ethnicity, smoking status, Charlson score, and rurality, findings were similar for 21 to 29 year old; Black women were screened less than White women [RR = 0.73 (0.67-0.79)]; current smokers [RR = 0.9 (0.85-0.94)] and past smokers [RR = 1.12 (1.06-1.17)] were screened less than never smokers. For 30 to 65 year old, Black women were screened less than White women [RR = 0.83 (0.79-0.88)]; current smokers [RR = 0.8 (0.78-0.81)] and past smokers [RR = 0.95 (0.93-0.96)] were screened less than never smokers. CONCLUSIONS Screening rates remained below the Healthy People 2030 goal of 79.2% over time, particularly for younger Black women and current smokers, with minimal use of primary HPV screening.
Collapse
Affiliation(s)
- Katie M. Ehman
- Department of Family Medicine, Mayo Clinic, Rochester, MN, USA
| | - Gregory D. Jenkins
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Jessica A. Grimm
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Julie A. Maxson
- Department of Family Medicine, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
3586
|
Huang S, Lai H, Pan X, Lin Q, Qin Y, Liu F, Fang M, Huang W, Wei C. Development and Validation of a Nomogram for Predicting Survival Based on Ferritin and Transferrin Ratio in Breast Cancer Patients. Cancer Control 2024; 31:10732748241261553. [PMID: 38850515 PMCID: PMC11162600 DOI: 10.1177/10732748241261553] [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: 11/24/2023] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Our objective is to develop a predictive model utilizing the ferritin and transferrin ratio (FTR) and clinical factors to forecast overall survival (OS) in breast cancer (BC) patients. METHODS We conducted a retrospective analysis of clinical data from 2858 BC patients diagnosed between 2013 and 2021. Subsequently, the cohort of 2858 BC patients underwent random assignment into distinct subsets: a training cohort comprising 2002 patients and a validation cohort comprising 856 patients, maintaining a proportional ratio of 7:3. Employing multivariable Cox regression analysis within the training cohort, we derived a prognostic nomogram. The predictive performance was assessed using calibration curves, C-index, and decision curve analysis. RESULTS The final prognostic model included the TNM stage, subtype, hemoglobin levels, and the ferritin-transferrin ratio. The nomogram achieved a C-index of .794 (95% CI: .777-.810). The nomogram demonstrated superior predictive accuracy for OS at 3, 5, and 7 years for BC, with area under the time-dependent curves of .812, .782, and .773, respectively. These values notably outperformed those of the conventional TNM stage. Decision curve analysis reaffirmed the greater net benefit of our nomogram compared to the TNM stage. These findings were subsequently validated in the independent validation cohort. CONCLUSION The FTR-based prognostic model may predict a patient's OS better than the TNM stage in a clinical setting. The nomogram can provide an early, affordable, and reliable tool for survival prediction, as well as aid clinicians in treatment option-making and prognosis evaluation. However, further multi-center prospective trials are required to confirm the reliability of the existing nomogram.
Collapse
Affiliation(s)
- Shuqing Huang
- Department of State-owned Asset Supervision and Administration Office, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Hao Lai
- Department of Guangxi Clinical Research Center for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaolan Pan
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiumei Lin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yuling Qin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fengfei Liu
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Min Fang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Wencheng Huang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Caibiao Wei
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
3587
|
Chen Y, Liu S, Tan S, Zheng Y, Chen Y, Yang C, Lin S, Mi Y, Li W. KRAS mutations promote the intratumoral colonization of enterotoxigenic bacteroides fragilis in colorectal cancer through the regulation of the miRNA3655/SURF6/IRF7/IFNβ axis. Gut Microbes 2024; 16:2423043. [PMID: 39523457 PMCID: PMC11556274 DOI: 10.1080/19490976.2024.2423043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
KRAS mutations are associated with poor prognosis in colorectal cancer (CRC). Although the association between the gut microbiota and CRC has been extensively documented, it is unclear whether KRAS mutations can regulate the gut microbiota. Metagenomics has identified changes in the diversity of the gut microbiota in CRC due to KRAS mutations. Specifically, KRAS mutations positively correlate with the abundance of the bacteroides. Understanding how to regulate the classic carcinogenic bacterium within the bacteroides, such as enterotoxigenic bacteroides fragilis (ETBF), to enhance treatment efficacy of tumors is a key focus of research. Mechanistically, we found that the reduction of miR3655 is indispensable for KRAS mutation-promoted proliferation of CRC and the abundance of ETBF. miR3655 targets SURF6 to inhibit its transcription. Further transcriptomic sequencing revealed that SURF6 promotes intratumoral colonization of ETBF in CRC by inhibiting the nuclear translocation and transcription levels of the IRF7, affecting the activation of the IFNβ promoter. Regulating miR3655 and SURF6 can promote IFNβ secretion in CRC, directly killing ETBF. These data indicate that KRAS mutations affect the intratumoral colonization of ETBF in CRC through the miR3655/SURF6/IRF7/IFNβ axis. This provides new potential strategies for treating CRC associated with KRAS mutations or high levels of ETBF.
Collapse
Affiliation(s)
- Yizhen Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian, China
| | - Shaolin Liu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian, China
| | - Song Tan
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian, China
| | - Yuanyuan Zheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Geriatric Medicine, Fujian Key Laboratory of Geriatrics Diseases, Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian, China
| | - Yifan Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian, China
| | - Changshun Yang
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian, China
| | - Shengtao Lin
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian, China
| | - Yulong Mi
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian, China
| | - Weihua Li
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian, China
| |
Collapse
|
3588
|
Yasin JA, Odat RM, Qtaishat FA, Tamimi MAA, Alsufi MI, Younis OM, Alkuttob LA, Saeed A. The Prognostic Significance of NEDD9 Expression in Human Cancers: A Systematic Review, Meta-Analysis, and Omics Exploration. Technol Cancer Res Treat 2024; 23:15330338241297597. [PMID: 39540210 PMCID: PMC11561999 DOI: 10.1177/15330338241297597] [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: 11/16/2024] Open
Abstract
BACKGROUND Neural precursor cell expressed developmentally downregulated 9 (NEDD9) is considered an important factor in the progression of cancer, acting as a modulator of cellular migration, adhesion, and metastatic potential. Its significance as a prognostic factor, however, remains unclear, which necessitated a comprehensive review and meta-analysis. METHODS Our study followed the PRISMA guidelines, analyzing studies from major databases including PubMed, Embase, and Cochrane. Our eligibility criteria included studies evaluating NEDD9 expression in relation to cancer prognosis and outcomes such as overall survival (OS), progression-free survival (PFS), disease-free Survival (DFS), recurrence-free survival (RFS), and cancer-specific survival (CSS). We used random-effects and fixed-effect models for meta-analysis, and we validated our findings by comparative analysis using data from external cohorts like The Cancer Genome Atlas (TCGA). RESULTS The analysis of 27 studies with 3915 patients demonstrated a significant relationship between NEDD9 expression and poor OS as indicated by the pooled meta-analysis outcome across all included cancers (HR: 1.81, 95% CI: 1.38-2.37). A significant effect on PFS/DFS/RFS/CSS was also found (HR: 2.14, 95% CI: 1.42-3.23). Variations in survival across different types of cancer were indicated by subgroup analysis. NEDD9 expression was correlated with various immune cells across cancer types according to immune infiltration analysis. Protein-protein interaction (PPI) analysis indicated significant interactions involving NEDD9, suggesting mechanisms which influence tumor behavior and response to treatment. CONCLUSION Our results suggest that NEDD9 is a significant prognostic marker in several human cancers. As a result of its central role in cancer progression and prognosis, it presents a promising target for therapeutic interventions. Our study highlights the importance of further research into the biology of NEDD9 and its therapeutic potential.
Collapse
Affiliation(s)
- Jehad A. Yasin
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Ramez M. Odat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | | | | | | | | | | | - Anwaar Saeed
- Division of Hematology & Oncology, Department of Medicine, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15232, USA
| |
Collapse
|
3589
|
Coyne B, Saravanakumar S, Murillo-Chavez F, Chengappa NK, Ihsan R, Chaudhry BA. Colorectal Carcinoma and Gluteal Abscesses in the Background of a Gait Disturbance Presentation. Case Rep Oncol 2024; 17:1239-1245. [PMID: 39469251 PMCID: PMC11517711 DOI: 10.1159/000541693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 09/20/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction Profuse diarrhea and abdominal discomfort are well-documented symptoms of patients with known colorectal cancer. It is much less common for these patients to present with a chief complaint of gait disturbance and rhabdomyolysis. We present a case of incidentally discovered colorectal carcinoma in a patient who was initially evaluated for progressive weakness and recurrent falls. Case Presentation A 51-year-old man was admitted to our department for management of rhabdomyolysis in the setting of progressive lower extremity weakness and mechanical falls. He developed abdominal discomfort and bowel changes during his admission, and after further investigation, he was found to have a rectal polyp positive for invasive adenocarcinoma, as well as multiple gluteal abscesses. Workup for metastasis, mutations, and oncogenic biomarkers was unremarkable. Conclusion This case is a demonstration of a medically complex patient presentation compounded by multifactorial processes. Future providers may take note that an initial absence of classic gastrointestinal (GI) symptoms does not necessarily rule out underlying GI cancer. Instead, the initial presentation of colorectal adenocarcinoma may manifest with paraneoplastic versus incidental progressive proximal limb weakness prior to GI symptoms such as diarrhea. Additionally, our report demonstrates a case of possible paraneoplastic gluteal abscesses, which may have further contributed to the patient's gait disturbance. However, it is unclear as to whether our patient's various symptoms were directly linked to one another, or if they were incidental co-presentations.
Collapse
Affiliation(s)
- Brendan Coyne
- George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Sanjeev Saravanakumar
- George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | | | | | - Rida Ihsan
- Department of Internal Medicine, Sinai Hospital, Baltimore, MD, USA
| | | |
Collapse
|
3590
|
Fullwood D, Fallon E, Bolajoko O, Gunderson J, Ramirez-Rivera CO, Pressey S, Nosbush K, Young ME, Krieger J, Asiedu G, Odedina FT. From Unheard to Empowered: Utilizing Communication Accommodation Theory to Address Black Men's With Prostate Cancer Healthcare Preferences. Cancer Control 2024; 31:10732748241293987. [PMID: 39865591 PMCID: PMC11489940 DOI: 10.1177/10732748241293987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/12/2024] [Accepted: 10/08/2024] [Indexed: 01/28/2025] Open
Abstract
OBJECTIVES Communication barriers, such as channels, comfort, and location, can negatively impact Black prostate cancer survivors' experiences and health outcomes after treatment. Addressing these barriers promotes a survivor-centric approach that views survivors as active partners in their care. This study explored the communication preferences of Black prostate cancer survivors, focusing on preferred channels, sources, and locations for enhanced quality of life. DESIGN Using grounded theory methodology, we conducted engaged in-depth interviews with 7 Black prostate cancer survivors, centering on their post-treatment experiences. We analyzed participant transcripts to develop codes and themes. Communication accommodation theory (CAT) was applied to examine how survivors adapt their communication styles to navigate barriers and improve their quality of life. CAT helped identify key patterns of survivors' convergence and divergence in their communication with healthcare providers, crucial for developing survivor-centric communication strategies. RESULTS The analysis revealed the importance of acknowledging cultural and racial perspectives in post-treatment survivorship. Themes extracted from the transcripts included communication barriers, preferences for information channels, and favored locations for sharing information. Survivors expressed a preference for a balance between face-to-face interaction and telehealth, with environments that provide safety and support playing a key role in encouraging vulnerability and open dialogue. Inclusive healthcare practices were seen as essential in empowering Black survivors to engage in meaningful communication without feeling silenced or undervalued, thus fostering a dignified quality of life. CONCLUSION Tailoring post-treatment communication practices to incorporate cultural sensitivity, inclusive healthcare, and safe spaces is pivotal to supporting Black survivors. These findings provide a pathway to improving survivors' experiences and empower them to actively shape their survivorship journey. Moreover, they highlight Black men's need for trustworthy communication with healthcare providers and peers during the post-treatment period, emphasizing the importance of direct, personalized interactions.
Collapse
Affiliation(s)
- Dottington Fullwood
- Cancer Prevention, Survivorship and Care Delivery (CPSCD) Research Program, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, USA
- iCCaRE for Black Men Consortium, Mayo Clinic, Jacksonville, FL, USA
| | - Elizabeth Fallon
- Department of Education, Mayo Clinic, College of Medicine and Science Rochester, Rochester, MN, USA
| | - Opeyemi Bolajoko
- Cancer Prevention, Survivorship and Care Delivery (CPSCD) Research Program, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, USA
- iCCaRE for Black Men Consortium, Mayo Clinic, Jacksonville, FL, USA
| | | | - Caleb O. Ramirez-Rivera
- Cancer Prevention, Survivorship and Care Delivery (CPSCD) Research Program, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, USA
- iCCaRE for Black Men Consortium, Mayo Clinic, Jacksonville, FL, USA
| | - Shannon Pressey
- Cancer Prevention, Survivorship and Care Delivery (CPSCD) Research Program, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, USA
- iCCaRE for Black Men Consortium, Mayo Clinic, Jacksonville, FL, USA
| | - Kelly Nosbush
- Cancer Prevention, Survivorship and Care Delivery (CPSCD) Research Program, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, USA
| | - Mary Ellen Young
- iCCaRE for Black Men Consortium, Mayo Clinic, Jacksonville, FL, USA
| | - Janice Krieger
- College of Medicine and Science, Mayo Clinic, Rochester, MN, USA
| | - Gladys Asiedu
- College of Medicine and Science, Mayo Clinic, Rochester, MN, USA
| | - Folakemi T. Odedina
- Cancer Prevention, Survivorship and Care Delivery (CPSCD) Research Program, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, USA
- iCCaRE for Black Men Consortium, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
3591
|
Tihagam RD, Bhatnagar S. Detection Methods for Epigenetic Mechanisms in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1465:99-103. [PMID: 39586996 DOI: 10.1007/978-3-031-66686-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Epigenetic dysregulation of gene expression is central to breast cancer initiation, progression, and treatment response. These alterations include histone modifications, DNA methylation, and expression of noncoding RNA. Technological advancements have improved our ability to identify histone modifications, DNA methylation patterns and provided critical insights into the epigenetic regulation of gene expression and its role in breast oncogenesis. The epigenetic profiles of healthy and breast cancer tissues revealed several diagnostic and prognostic biomarkers. In this article, we review the methodologies commonly used to study tumor-associated histone modifications and DNA methylation changes in breast cancer, highlighting their principles, applications, and advancements, such as chromatin immunoprecipitation (ChIP), bisulfite conversion of DNA, next-generation sequencing (NGS), and chromatin immunoprecipitation sequencing (ChIP-seq).
Collapse
Affiliation(s)
- Rachisan Djiake Tihagam
- Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Davis, CA, USA
| | - Sanchita Bhatnagar
- Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Davis, CA, USA.
| |
Collapse
|
3592
|
Carroll EF, Rogers C, Summerside M, Cortina CS. Breast care considerations for transgender and gender-diverse patients. WOMEN'S HEALTH (LONDON, ENGLAND) 2024; 20:17455057241289706. [PMID: 39382481 PMCID: PMC11465296 DOI: 10.1177/17455057241289706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024]
Abstract
Transgender and gender-diverse (TGD) persons represent a small but growing population in the United States. Accessing inclusive, equitable, and evidence-based healthcare remains a challenge for this patient population. Many TGD persons seek gender-affirming care, including gender-affirming hormonal therapy (GAHT) and gender-affirming surgery (GAS), to help ameliorate the physical and mental aspects of their gender incongruence. Both GAHT and GAS induce clinically important histopathologic and anatomic changes in breast tissue. Consequently, breast care in TGD persons has become an increasingly recognized topic of importance in gender-affirming care. However, there remains a scarce but growing base of literature specifically addressing the unique healthcare needs of breast care in TGD patients. This article will review how to establish trusting patient-provider relationships for TGD patients, gender inclusivity in breast clinics and imaging centers, the influence of GAHT and GAS on breast tissue, breast cancer screening recommendations and barriers, and breast cancer risk and treatment considerations in TGD persons.
Collapse
Affiliation(s)
- Evelyn F Carroll
- Division of Breast Imaging and Intervention, Department of Radiology, Mayo Clinic, Rochester, MN, USA
- Division of Emergency and Hospital Radiology, Department of Radiology, Mayo Clinic, Rochester, MN, 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
|
3593
|
Valtingojer I, Lièvre S, Bordes P, Paranjpe K, Thompson W, Shah S, Fantin V, Jacquemet-Ross W, Adamson PC. Collaborative Innovations in Childhood Cancer Therapies. Handb Exp Pharmacol 2024; 286:33-50. [PMID: 39177748 DOI: 10.1007/164_2024_725] [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: 08/24/2024]
Abstract
The outcome for children with cancer has improved significantly over the past 60 years, with more than 80% of patients today becoming 5-year survivors. Despite this progress, cancer remains the leading cause of death from disease in children in the United States and Europe, with significant short- and long-term toxicity of treatment continuing to impact most children. While the past 15 years have witnessed dramatic scientific innovation for certain cancers in adult patients, pediatric cancer treatment innovation lags increasingly behind. To help bridge the adult-pediatric therapeutic development gap, collaborative efforts are essential among stakeholders within and outside the pediatric oncology community. Prioritizing collaboration in areas such as cancer characterization, target identification and validation, drug discovery, and approaches to currently "undruggable" targets is imperative to improving the outcomes for children with cancer.
Collapse
Affiliation(s)
| | - Sasha Lièvre
- Department of Oncology, R&D, Sanofi, Vitry-sur-Seine, France
| | - Philippe Bordes
- Department of Alliance Management and Public Private Partnerships, Sanofi, Gentilly, France
| | - Krupa Paranjpe
- Department of Oncology Medical Affairs, Sanofi, Cambridge, MA, USA
| | | | - Sachin Shah
- Department of Oncology Medical Affairs, Sanofi, Cambridge, MA, USA
| | | | | | | |
Collapse
|
3594
|
Dizon DS, Kamal AH. Cancer statistics 2024: All hands on deck. CA Cancer J Clin 2024; 74:8-9. [PMID: 38230825 DOI: 10.3322/caac.21824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2024] Open
Affiliation(s)
- Don S Dizon
- Lifespan Cancer Institute, Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center, Brown University, Providence, Rhode Island, USA
| | | |
Collapse
|
3595
|
Jia D, Kuang Z, Wang L. The role of microbial indole metabolites in tumor. Gut Microbes 2024; 16:2409209. [PMID: 39353090 PMCID: PMC11445886 DOI: 10.1080/19490976.2024.2409209] [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: 07/13/2024] [Revised: 08/30/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024] Open
Abstract
The gut microbiota can produce a variety of microbial-derived metabolites to influence tumor development. Tryptophan, an essential amino acid in the human body, can be converted by microorganisms via the indole pathway to indole metabolites such as Indole-3-Lactic Acid (ILA), Indole-3-Propionic Acid (IPA), Indole Acetic Acid (IAA) and Indole-3-Aldehyde (IAld). Recent studies have shown that indole metabolites play key roles in tumor progression, and they can be used as adjuvant regimens for tumor immunotherapy or chemotherapy. Here, we summarize recent findings on the common microbial indole metabolites and provide a review of the mechanisms of different indole metabolites in the tumor microenvironment. We further discuss the limitations of current indole metabolite research and future possibilities. It is expected that microbial indole metabolites will provide new strategies for clinical therapy.
Collapse
Affiliation(s)
- Dingjiacheng Jia
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Zheng Kuang
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Liangjing Wang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
3596
|
Thompson T, Lero CM, Levitan J, Bradley CD, James AS, Heiden-Rootes K, Sargent E, Johnson LP. Insights About Dyadic Cancer Survivorship Interventions for Black Women and Their Caregivers: A Rapid Qualitative Analysis of Collaborator Perspectives. Cancer Control 2024; 31:10732748241305583. [PMID: 39672808 PMCID: PMC11645766 DOI: 10.1177/10732748241305583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/25/2024] [Accepted: 11/19/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND Breast cancer patients and their informal caregivers often report unmet psychosocial, relational, and physical health needs. Dyadic interventions may improve patient and caregiver outcomes, but few have been integrated into clinical care or designed for Black breast cancer patients and their female caregivers. We used the Health Equity Implementation Framework to design for dissemination by identifying facilitators and barriers to implementing a dyadic survivorship intervention delivered via video teleconferencing (e.g., Zoom). METHODS We conducted semi-structured interviews with a purposive sample of 18 collaborators with roles at a comprehensive cancer center and community organizations to understand facilitators and barriers to implementing a dyadic intervention for Black breast cancer patients and their caregivers. We used rapid qualitative analysis (templated summaries synthesized in matrices) to conduct a directed content analysis. We identified patterns in responses to interview guide questions and developed cross-cutting themes. RESULTS Collaborators' roles fell into four domains: patient-facing mental health (33%), patient-facing physical health (28%), research/administration (28%), and cancer-focused community groups (11%). Participants were supportive of a dyadic intervention for Black women with breast cancer and female caregivers. Collaborators noted that psychosocial care at the cancer center was already being delivered via Zoom and saw benefits to including caregivers. Overarching themes include the need to address gaps in care (for caregivers, Black women, and long-term breast cancer survivors); the importance of representation in building trust and mitigating stigma; and the challenges within the healthcare system around providing care services to dyads. CONCLUSION Application of these findings can help address the challenges of implementing a dyadic survivorship intervention for Black women with breast cancer and their caregivers in clinical settings. Expanding models such as the Health Equity Implementation Framework to include caregivers may help focus dissemination and implementation efforts on both members of a dyad and improve outcomes for both.
Collapse
Affiliation(s)
- Tess Thompson
- University of North Carolina at Chapel Hill, School of Social Work, Chapel Hill, NC, USA
| | - Christi M. Lero
- Washington University in St. Louis, Brown School of Social Work, St. Louis, MO, USA
| | - Julia Levitan
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, Chapel Hill, NC, USA
| | - Cory D. Bradley
- Northwestern University, Feinberg School of Medicine, Department of Medical Social Sciences, Chicago, IL, USA
| | - Aimee S. James
- Washington University School of Medicine, Department of Surgery, St. Louis, MO, USA
| | | | - Emani Sargent
- Washington University in St. Louis, Brown School of Social Work, St. Louis, MO, USA
| | - LaShaune P. Johnson
- University of Houston, Department of Health Systems and Population Health Sciences, Houston, TX, USA
| |
Collapse
|
3597
|
Chan KH, Clavijo ND, Ayala G, Hall R, Wray C, Cen P. Double Hit in Clear-Cell Renal Cell Carcinoma With Germline Pathogenic ATM Mutation and Somatic VHL Mutation. J Investig Med High Impact Case Rep 2024; 12:23247096241286370. [PMID: 39369320 PMCID: PMC11457202 DOI: 10.1177/23247096241286370] [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: 07/30/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 10/07/2024] Open
Abstract
While renal cell carcinoma (RCC) is often linked to smoking, obesity, and hypertension, hereditary forms also account for about 3% of RCC cases. Notably, NCCN guidelines identify 7 major hereditary syndromes associated with an increased RCC risk. Inherited mutations in DNA repair genes, such as ATM, BRCA, and TP53, significantly increase the risk of various cancers. Biallelic pathogenic mutations in ATM cause Ataxia-Telangiectasia (A-T) syndrome, while heterozygous germline pathogenic ATM mutations, present in about 1% of the population, also elevate cancer risk. RCC has not traditionally been associated with germline pathogenic ATM mutations, only limited retrospective analyses have identified such mutations. This case report presents a 68-year-old woman with a germline pathogenic ATM mutation (c.8786+1 G>A) who developed high-risk clear cell RCC followed by an acquired somatic VHL mutation in RCC and a 3-cm serous cystadenoma, illustrating the double-hit phenomenon. Her brother, who shares the same germline pathogenic mutation, was diagnosed with pancreatic cancer and prostate cancer. This case highlights the potential use for enhanced screening protocols for RCC in patients who have germline pathogenic ATM mutations and the importance of research in targeted treatments for tumors driven by dual genetic mechanisms. Increased awareness and vigilant screening for RCC are crucial in managing hereditary cancer syndromes effectively.
Collapse
Affiliation(s)
- Kok Hoe Chan
- The University of Texas Health Science Center at Houston, USA
| | | | - Gustavo Ayala
- The University of Texas Health Science Center at Houston, USA
| | - Ryan Hall
- The University of Texas Health Science Center at Houston, USA
| | - Curtis Wray
- The University of Texas Health Science Center at Houston, USA
| | - Putao Cen
- The University of Texas Health Science Center at Houston, USA
| |
Collapse
|
3598
|
Guefack MGF, Bhatnagar S. Advances in Epigenetic Therapeutics for Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1465:89-97. [PMID: 39586995 DOI: 10.1007/978-3-031-66686-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
The epigenetic deregulations correlate with tumorigenesis, resistance to therapy, and metastasis of breast cancer cells. Given the predominance of aberrant epigenomic mechanisms, there is a growing emphasis on targeting epigenetic mechanisms for breast cancer therapeutic development. Selective inhibitors of epigenetic enzymes and the combined approach of epigenetic therapies with chemotherapies or hormone therapies in the treatment of breast cancer represent promising therapeutic strategies. In this chapter, we review the targeting of epigenetic mechanisms and highlight current epigenetic research in the development of breast cancer therapy.
Collapse
Affiliation(s)
- Michel-Gael F Guefack
- Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Davis, CA, USA
| | - Sanchita Bhatnagar
- Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Davis, CA, USA.
| |
Collapse
|
3599
|
Greene M, Pew T, Le Q, Philp A, Johnson WK, Ozbay AB, Kisiel J, Dore M, Ebner D, Fendrick AM, Limburg P. Member Adherence to a Health Insurer-Sponsored Gap Closure Program Using Multi-Target Stool DNA Test for Colorectal Cancer Screening. J Prim Care Community Health 2024; 15:21501319241305958. [PMID: 39655481 PMCID: PMC11629422 DOI: 10.1177/21501319241305958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVES To describe member adherence to a mail-based, health insurer-sponsored gap closure program for colorectal cancer (CRC) screening using multi-target stool DNA (mt-sDNA; Cologuard®) tests. METHODS Combined patient data from Exact Sciences Laboratories LLC and data from mass-mailed mt-sDNA orders placed by a large Medicare Advantage Insurance Plan were analyzed (03/01/2023-06/30/2023). Adherence and time to test return were the primary and secondary outcomes, respectively. Their association with patient characteristics was evaluated using multivariable regression. RESULTS Among the 3201 member-patients included (86.6% aged 65-75 years; 58.7% female), adherence rate was 49.4%, and mean time to test return was 25.5 days. After multivariable adjustment, the odds of test return were significantly higher among 65- to 75-year-olds (odds ratio [OR] = 1.59 relative to 45- to 64-year-olds), those living in small towns (OR = 1.43 relative to metropolitan-located individuals), and with digital outreach via both SMS and email (OR = 4.31 relative to no digital outreach). Time to test return was shorter in 65- to 75-year-olds than in 45- to 64-year-olds and was not associated with other patient characteristics. CONCLUSIONS Mass-mailed mt-sDNA tests for CRC screening were associated with an overall adherence rate of about 50% in this Medicare Advantage population, with higher likelihood of test return among patients receiving digital outreach.
Collapse
Affiliation(s)
| | - Timo Pew
- Exact Sciences Corporation, Madison, WI, USA
| | - Quang Le
- Exact Sciences Corporation, Madison, WI, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
3600
|
Petrella F, Cara A, Cassina EM, Faverio P, Franco G, Libretti L, Pirondini E, Raveglia F, Sibilia MC, Tuoro A, Vaquer S, Luppi F. Evaluation of preoperative cardiopulmonary reserve and surgical risk of patients undergoing lung cancer resection. Ther Adv Respir Dis 2024; 18:17534666241292488. [PMID: 39455414 PMCID: PMC11523151 DOI: 10.1177/17534666241292488] [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: 05/06/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Lung cancer represents the second most frequent neoplasm and the leading cause of neoplastic death among both women and men, causing almost 25% of all cancer deaths. Patients undergoing lung resection-both for primary and secondary tumors-require careful preoperative cardiopulmonary functional evaluation to confirm the safety of the planned resection, to assess the maximum tolerable volume of resection or to exclude surgery, thus shifting the therapeutic approach toward less invasive options. Cardiopulmonary reserve, pulmonary lung function and mechanical respiratory function represent the cornerstones of preoperative assessment of patients undergoing major lung resection. Spirometry with carbon monoxide diffusing capacity, split function tests, exercise tests and cardiologic evaluation are the gold standard instruments to safely assess the entire cardiorespiratory function before pulmonary resection. Although pulmonary mechanical and parenchymal function, together with cardiorespiratory compliance represent the mainstay of preoperative evaluation in thoracic surgery, the variables that are responsible for fitness in patients who have undergone lung resection have expanded and are being continually investigated. Nevertheless, because of the shift to older patients who undergo lung resection, a global approach is required, taking into consideration variables like frailty status and likelihood of postoperative functional deterioration. Finally, the decision to go ahead with surgery in fragile patients being consideredfor lung resection should be evaluated in a multispecialty preoperative discussion to provide a personalized risk stratification. The aim of this review is to focus on preoperative evaluation of cardiopulmonary reserve and surgical risk stratification of patients candidate for lung cancer resection. It does so by a literature search of clinical guidelines, expert consensus statements, meta-analyses, clinical recommendations, book chapters and randomized trials (1980-2022).
Collapse
Affiliation(s)
- Francesco Petrella
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, Monza (MB) 20090, Italy
| | - Andrea Cara
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Enrico Mario Cassina
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Paola Faverio
- Division of Respiratory Disease, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Giovanni Franco
- Division of Respiratory Disease, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Lidia Libretti
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Emanuele Pirondini
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Federico Raveglia
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Maria Chiara Sibilia
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Antonio Tuoro
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Sara Vaquer
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Fabrizio Luppi
- Division of Respiratory Disease, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|