3501
|
Dosunmu GT, Shergill A. Colorectal Cancer: Genetic Underpinning and Molecular Therapeutics for Precision Medicine. Genes (Basel) 2024; 15:538. [PMID: 38790167 PMCID: PMC11120657 DOI: 10.3390/genes15050538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/16/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Colorectal cancer (CRC) accounts for about 10% of all cancer cases and 9% of cancer-related deaths globally. In the United States alone, CRC represents approximately 12.6% of all cancer cases, with a mortality rate of about 8%. CRC is now the first leading cause of cancer death in men younger than age 50 and second in women younger than age 50. This review delves into the genetic landscape of CRC, highlighting key mutations and their implications in disease progression and treatment. We provide an overview of the current and emerging therapeutic strategies tailored to individual genomic profiles.
Collapse
Affiliation(s)
| | - Ardaman Shergill
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA;
| |
Collapse
|
3502
|
Karaoğlan BB, Ürün Y. Unveiling the Role of Human Papillomavirus in Urogenital Carcinogenesis a Comprehensive Review. Viruses 2024; 16:667. [PMID: 38793549 PMCID: PMC11125962 DOI: 10.3390/v16050667] [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: 02/29/2024] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 05/26/2024] Open
Abstract
Human papillomavirus (HPV), an oncogenic DNA virus, is the most common sexually transmitted virus and significant public health concern globally. Despite the substantial prevalence of HPV infection among men, routine testing remains elusive due to the lack of approved HPV tests and the complexity of detection methods. Various studies have explored the link between HPV and genitourinary cancers, revealing different associations influenced by geographic variation, histological subtype and methodological differences. These findings underscore the importance of further research to elucidate the role of HPV in male urogenital cancers. This comprehensive review delves into the intricate relationship between HPV and male genitourinary cancers, shedding light on the virus's oncogenic mechanisms and its reported prevalence. A deeper understanding of HPV's implications for male health is essential for advancing public health initiatives and reducing the burden of urogenital cancers worldwide.
Collapse
Affiliation(s)
- Beliz Bahar Karaoğlan
- Department of Medical Oncology, Ankara University Faculty of Medicine, 06620 Ankara, Türkiye;
- Faculty of Medicine, Department of Internal Medicine, Division of Internal Medicine, Ankara University Cancer Research Institute, 06620 Ankara, Türkiye
| | - Yüksel Ürün
- Department of Medical Oncology, Ankara University Faculty of Medicine, 06620 Ankara, Türkiye;
- Faculty of Medicine, Department of Internal Medicine, Division of Internal Medicine, Ankara University Cancer Research Institute, 06620 Ankara, Türkiye
| |
Collapse
|
3503
|
Adamopoulos C, Papavassiliou KA, Poulikakos PI, Papavassiliou AG. RAF and MEK Inhibitors in Non-Small Cell Lung Cancer. Int J Mol Sci 2024; 25:4633. [PMID: 38731852 PMCID: PMC11083651 DOI: 10.3390/ijms25094633] [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: 02/29/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Lung cancer, despite recent advancements in survival rates, represents a significant global health burden. Non-small cell lung cancer (NSCLC), the most prevalent type, is driven largely by activating mutations in Kirsten rat sarcoma viral oncogene homologue (KRAS) and receptor tyrosine kinases (RTKs), and less in v-RAF murine sarcoma viral oncogene homolog B (BRAF) and mitogen-activated protein-kinase kinase (MEK), all key components of the RTK-RAS-mitogen-activated protein kinase (MAPK) pathway. Learning from melanoma, the identification of BRAFV600E substitution in NSCLC provided the rationale for the investigation of RAF and MEK inhibition as a therapeutic strategy. The regulatory approval of two RAF-MEK inhibitor combinations, dabrafenib-trametinib, in 2017, and encorafenib-binimetinib, in 2023, signifies a breakthrough for the management of BRAFV600E-mutant NSCLC patients. However, the almost universal emergence of acquired resistance limits their clinical benefit. New RAF and MEK inhibitors, with distinct biochemical characteristics, are in preclinical and clinical development. In this review, we aim to provide valuable insights into the current state of RAF and MEK inhibition in the management of NSCLC, fostering a deeper understanding of the potential impact on patient outcomes.
Collapse
Affiliation(s)
- Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Poulikos I. Poulikakos
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
3504
|
Thoidingjam S, Sriramulu S, Hassan O, Brown SL, Siddiqui F, Movsas B, Gadgeel S, Nyati S. BUB1 inhibition sensitizes lung cancer cell lines to radiotherapy and chemoradiotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590355. [PMID: 38712071 PMCID: PMC11071420 DOI: 10.1101/2024.04.19.590355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Background Lung cancer is a major public health concern, with high incidence and mortality. Despite advances in targeted therapy and immunotherapy, microtubule stabilizers (paclitaxel, docetaxel), DNA intercalating platinum drugs (cisplatin) and radiation therapy continue to play a critical role in the management of locally advanced and metastatic lung cancer. Novel molecular targets would provide opportunities for improving the efficacies of radiotherapy and chemotherapy. Hypothesis We hypothesize that BUB1 (Ser/Thr kinase) is over-expressed in lung cancers and that its inhibition will sensitize lung cancers to chemoradiation. Methods BUB1 inhibitor (BAY1816032) was combined with platinum (cisplatin), microtubule poison (paclitaxel), a PARP inhibitor (olaparib) and radiation in cell proliferation and radiation sensitization assays. Biochemical and molecular assays were used to evaluate their impact on DNA damage signaling and cell death mechanisms. Results BUB1 expression assessed by immunostaining of lung tumor microarrays (TMAs) confirmed higher BUB1 expression in NSCLC and SCLC compared to that of normal tissues. BUB1 overexpression in lung cancer tissues correlated directly with expression of TP53 mutations in non-small cell lung cancer (NSCLC). Elevated BUB1 levels correlated with poorer overall survival in NSCLC and small cell lung cancer (SCLC) patients. A BUB1 inhibitor (BAY1816032) synergistically sensitized lung cancer cell lines to paclitaxel and olaparib. Additionally, BAY1816032 enhanced cell killing by radiation in both NSCLC and SCLC. Molecular changes following BUB1 inhibition suggest a shift towards pro-apoptotic and anti-proliferative states, indicated by altered expression of BAX, BCL2, PCNA, and Caspases 9 and 3. Conclusion A direct correlation between BUB1 protein expression and overall survival was shown. BUB1 inhibition sensitized both NSCLC and SCLC to various chemotherapies (cisplatin, paclitaxel) and targeted therapy (PARPi). Furthermore, we present the novel finding that BUB1 inhibition sensitized both NSCLC and SCLC to radiotherapy and chemoradiation. Our results demonstrate BUB1 inhibition as a promising strategy to sensitize lung cancers to radiation and chemoradiation therapies.
Collapse
|
3505
|
Gaebler D, Hachey SJ, Hughes CCW. Microphysiological systems as models for immunologically 'cold' tumors. Front Cell Dev Biol 2024; 12:1389012. [PMID: 38711620 PMCID: PMC11070549 DOI: 10.3389/fcell.2024.1389012] [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: 02/20/2024] [Accepted: 03/25/2024] [Indexed: 05/08/2024] Open
Abstract
The tumor microenvironment (TME) is a diverse milieu of cells including cancerous and non-cancerous cells such as fibroblasts, pericytes, endothelial cells and immune cells. The intricate cellular interactions within the TME hold a central role in shaping the dynamics of cancer progression, influencing pivotal aspects such as tumor initiation, growth, invasion, response to therapeutic interventions, and the emergence of drug resistance. In immunologically 'cold' tumors, the TME is marked by a scarcity of infiltrating immune cells, limited antigen presentation in the absence of potent immune-stimulating signals, and an abundance of immunosuppressive factors. While strategies targeting the TME as a therapeutic avenue in 'cold' tumors have emerged, there is a pressing need for novel approaches that faithfully replicate the complex cellular and non-cellular interactions in order to develop targeted therapies that can effectively stimulate immune responses and improve therapeutic outcomes in patients. Microfluidic devices offer distinct advantages over traditional in vitro 3D co-culture models and in vivo animal models, as they better recapitulate key characteristics of the TME and allow for precise, controlled insights into the dynamic interplay between various immune, stromal and cancerous cell types at any timepoint. This review aims to underscore the pivotal role of microfluidic systems in advancing our understanding of the TME and presents current microfluidic model systems that aim to dissect tumor-stromal, tumor-immune and immune-stromal cellular interactions in various 'cold' tumors. Understanding the intricacies of the TME in 'cold' tumors is crucial for devising effective targeted therapies to reinvigorate immune responses and overcome the challenges of current immunotherapy approaches.
Collapse
Affiliation(s)
- Daniela Gaebler
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Stephanie J. Hachey
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Christopher C. W. Hughes
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
- Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
3506
|
Lee YJ, Kim WR, Park EG, Lee DH, Kim JM, Shin HJ, Jeong HS, Roh HY, Kim HS. Exploring the Key Signaling Pathways and ncRNAs in Colorectal Cancer. Int J Mol Sci 2024; 25:4548. [PMID: 38674135 PMCID: PMC11050203 DOI: 10.3390/ijms25084548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent cancer to be diagnosed, and it has a substantial mortality rate. Despite numerous studies being conducted on CRC, it remains a significant health concern. The disease-free survival rates notably decrease as CRC progresses, emphasizing the urgency for effective diagnostic and therapeutic approaches. CRC development is caused by environmental factors, which mostly lead to the disruption of signaling pathways. Among these pathways, the Wingless/Integrated (Wnt) signaling pathway, Phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway, Mitogen-Activated Protein Kinase (MAPK) signaling pathway, Transforming Growth Factor-β (TGF-β) signaling pathway, and p53 signaling pathway are considered to be important. These signaling pathways are also regulated by non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). They have emerged as crucial regulators of gene expression in CRC by changing their expression levels. The altered expression patterns of these ncRNAs have been implicated in CRC progression and development, suggesting their potential as diagnostic and therapeutic targets. This review provides an overview of the five key signaling pathways and regulation of ncRNAs involved in CRC pathogenesis that are studied to identify promising avenues for diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Yun Ju Lee
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Woo Ryung Kim
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Eun Gyung Park
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Du Hyeong Lee
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Jung-min Kim
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Hae Jin Shin
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Hyeon-su Jeong
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (Y.J.L.); (W.R.K.); (E.G.P.); (D.H.L.); (J.-m.K.); (H.J.S.); (H.-s.J.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
| | - Hyun-Young Roh
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Heui-Soo Kim
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea;
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
3507
|
Zhang Y, Zhang Y, Song J, Cheng X, Zhou C, Huang S, Zhao W, Zong Z, Yang L. Targeting the "tumor microenvironment": RNA-binding proteins in the spotlight in colorectal cancer therapy. Int Immunopharmacol 2024; 131:111876. [PMID: 38493688 DOI: 10.1016/j.intimp.2024.111876] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/04/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
Colorectal cancer (CRC) is the third most common cancer and has the second highest mortality rate among cancers. The development of CRC involves both genetic and epigenetic abnormalities, and recent research has focused on exploring the ex-transcriptome, particularly post-transcriptional modifications. RNA-binding proteins (RBPs) are emerging epigenetic regulators that play crucial roles in post-transcriptional events. Dysregulation of RBPs can result in aberrant expression of downstream target genes, thereby affecting the progression of colorectal tumors and the prognosis of patients. Recent studies have shown that RBPs can influence CRC pathogenesis and progression by regulating various components of the tumor microenvironment (TME). Although previous research on RBPs has primarily focused on their direct regulation of colorectal tumor development, their involvement in the remodeling of the TME has not been systematically reported. This review aims to highlight the significant role of RBPs in the intricate interactions within the CRC tumor microenvironment, including tumor immune microenvironment, inflammatory microenvironment, extracellular matrix, tumor vasculature, and CRC cancer stem cells. We also highlight several compounds under investigation for RBP-TME-based treatment of CRC, including small molecule inhibitors such as antisense oligonucleotides (ASOs), siRNAs, agonists, gene manipulation, and tumor vaccines. The insights gained from this review may lead to the development of RBP-based targeted novel therapeutic strategies aimed at modulating the TME, potentially inhibiting the progression and metastasis of CRC.
Collapse
Affiliation(s)
- Yiwei Zhang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China; Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China; Queen Mary School, Nanchang University, 330006 Nanchang, China
| | - Yujun Zhang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China; Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China
| | - Jingjing Song
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China; Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China; School of Ophthalmology and Optometry of Nanchang University, China
| | - Xifu Cheng
- School of Ophthalmology and Optometry of Nanchang University, China
| | - Chulin Zhou
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Shuo Huang
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Wentao Zhao
- The 3rd Clinical Department of China Medical University, 10159 Shenyang, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China.
| | - Lingling Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China.
| |
Collapse
|
3508
|
Shen J, Sun H, Chu J, Gong X, Liu X. Cervicovaginal microbiota: a promising direction for prevention and treatment in cervical cancer. Infect Agent Cancer 2024; 19:13. [PMID: 38641803 PMCID: PMC11027553 DOI: 10.1186/s13027-024-00573-8] [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: 02/05/2024] [Accepted: 03/18/2024] [Indexed: 04/21/2024] Open
Abstract
Cervical cancer is a common malignancy in women, with high incidence rate and mortality. Persistent infection of high-risk human papillomavirus (HPV) is the most important risk factor for cervical cancer and precancerous lesions. Cervicovaginal microbiota (CVM) plays an essential role in the defense of HPV infections and prevention of subsequent lesions. Dominance of Lactobacillus is the key of CVM homeostasis, which can be regulated by host, exogenous and endogenous factors. Dysbiosis of CVM, including altered microbial, metabolic, and immune signatures, can contribute to persist HPV infection, leading to cervical cancer. However, there is no evidence of the causality between CVM and cervical cancer, and the underlying mechanism remains unexplored. Considering the close correlation between CVM dysbiosis and persistent HPV infection, this review will overview CVM, its role in cervical cancer development and related mechanisms, and the prospects for therapeutic applications.
Collapse
Affiliation(s)
- Jie Shen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 200003, Shanghai, China
| | - Hao Sun
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 200003, Shanghai, China
| | - Jing Chu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 200003, Shanghai, China
| | - Xiaodi Gong
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 200003, Shanghai, China.
| | - Xiaojun Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), 200003, Shanghai, China.
| |
Collapse
|
3509
|
Fernández-Sánchez J, Trujillo-Colmena D, Rodríguez-Castaño A, Lavín-Pérez AM, Del Coso J, Casado A, Collado-Mateo D. Effects of exercise on life satisfaction of people diagnosed with cancer: a systematic review and meta-analysis. Support Care Cancer 2024; 32:297. [PMID: 38637349 PMCID: PMC11026230 DOI: 10.1007/s00520-024-08486-3] [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: 12/13/2023] [Accepted: 04/03/2024] [Indexed: 04/20/2024]
Abstract
PURPOSE A cancer diagnosis is commonly associated with a decline in patient's life satisfaction and more pessimistic expectations about the future. The identification of strategies to improve life satisfaction in patients with cancer is of great interest to health practitioners since it may be associated with a better prognosis of cancer and higher survival rates. Previous meta-analyses and reviews concluded that exercise could significantly improve health-related quality of life in this population, but the effects of exercise on life satisfaction are still not well-known. This review aims to analyse the effects of exercise programs on life satisfaction in people with cancer and individuals who have overcome cancer. METHODS The present systematic review and meta-analysis followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. A thorough search of databases including Web of Science and PubMed/MEDLINE was carried out. Six studies (535 participants) in which the effect of an exercise program was compared to a non-exercise program control condition in patients with cancer were considered eligible. A subsequent meta-analysis was performed using the random effects model to calculate the standardized mean differences (SMD) and 95% confidence intervals (CI). RESULTS Exercise intervention improved satisfaction with life compared with a control condition (SMD = 1.28; p = 0.02 with a 95% CI of 0.22 to 2.34). CONCLUSION Exercise could be considered an effective tool to improve life satisfaction in patients with cancer. Hence, professionals might consider the possibility of integrating physical exercise into strategies aimed at enhancing the low life satisfaction often experienced by patients. PROSPERO CRD42023438146.
Collapse
Affiliation(s)
| | | | | | - Ana Myriam Lavín-Pérez
- Sport Sciences Research Centre, Rey Juan Carlos University, Fuenlabrada, Madrid, Spain.
- GO fitLAB, Ingesport, Madrid, Spain.
| | - Juan Del Coso
- Sport Sciences Research Centre, Rey Juan Carlos University, Fuenlabrada, Madrid, Spain
| | - Arturo Casado
- Sport Sciences Research Centre, Rey Juan Carlos University, Fuenlabrada, Madrid, Spain
| | - Daniel Collado-Mateo
- Sport Sciences Research Centre, Rey Juan Carlos University, Fuenlabrada, Madrid, Spain
| |
Collapse
|
3510
|
Fateeva A, Eddy K, Chen S. Current State of Melanoma Therapy and Next Steps: Battling Therapeutic Resistance. Cancers (Basel) 2024; 16:1571. [PMID: 38672652 PMCID: PMC11049326 DOI: 10.3390/cancers16081571] [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: 03/05/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Melanoma is the most aggressive and deadly form of skin cancer due to its high propensity to metastasize to distant organs. Significant progress has been made in the last few decades in melanoma therapeutics, most notably in targeted therapy and immunotherapy. These approaches have greatly improved treatment response outcomes; however, they remain limited in their abilities to hinder disease progression due, in part, to the onset of acquired resistance. In parallel, intrinsic resistance to therapy remains an issue to be resolved. In this review, we summarize currently available therapeutic options for melanoma treatment and focus on possible mechanisms that drive therapeutic resistance. A better understanding of therapy resistance will provide improved rational strategies to overcome these obstacles.
Collapse
Affiliation(s)
- Anna Fateeva
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
| | - Kevinn Eddy
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
| | - Suzie Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- U.S. Department of Veterans Affairs, New Jersey Health Care System, East Orange, NJ 07018, USA
| |
Collapse
|
3511
|
Tivadar BM, Dumitrascu T, Vasilescu C. A Glimpse into the Role and Effectiveness of Splenectomy for Isolated Metachronous Spleen Metastasis of Colorectal Cancer Origin: Long-Term Survivals Can Be Achieved. J Clin Med 2024; 13:2362. [PMID: 38673636 PMCID: PMC11050850 DOI: 10.3390/jcm13082362] [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: 03/06/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Background: Many papers exploring the role of resectioning metastases in colorectal cancer (CRC) have focused mainly on liver and lung sites, showing improved survival compared with non-resectional therapies. However, data about exceptional metastatic sites such as splenic metastases (SMs) are scarce. This paper aims to assess the role and effectiveness of splenectomy in the case of isolated metachronous SM of CRC origin. Methods: The patients' data were extracted after a comprehensive literature search through public databases for articles reporting patients with splenectomies for isolated metachronous SM of CRC origin. Potential predictors of survival were explored, along with demographic, diagnostic, pathology, and treatment data for each patient. Results: A total of 83 patients with splenectomies for isolated metachronous SM of CRC origin were identified. The primary CRC was at an advanced stage (Duke's C-70.3%) and on the left colon (45.5%) for most patients, while the median interval between CRC resection and SM was 24 months. The median overall survival after splenectomy was 84 months, and patients younger than 62 years presented statistically significantly worse overall survival rates than those ≥62 years old (p = 0.011). There was no significant impact on the long-term outcomes for factors including primary tumor location or adjuvant chemotherapy (p values ≥ 0.070, ns). Laparoscopic splenectomy was increasingly used in the last 20 years from 2002 (33.3% vs. 0%, p < 0.001). Conclusions: Splenectomy is the optimal treatment for patients with isolated metachronous SM of CRC, with the laparoscopic approach being increasingly used and having the potential to become a standard of care. Encouraging long-term survival rates were reported in the context of a multidisciplinary approach. Younger ages are associated with worse survival. Perioperative chemotherapy in the context of a patient diagnosed with SM of CRC origin appears to be a reasonable option, although the present study failed to show any significant impact on long-term survival.
Collapse
Affiliation(s)
| | - Traian Dumitrascu
- Department of General Surgery, Fundeni Clinical Institute, Carol Davila University of Medicine and Pharmacy, Fundeni Street No. 258, 022328 Bucharest, Romania; (B.M.T.); (C.V.)
| | | |
Collapse
|
3512
|
Vishwanath A, Krishna S, Manudhane AP, Hart PA, Krishna SG. Early-Onset Gastrointestinal Malignancies: An Investigation into a Rising Concern. Cancers (Basel) 2024; 16:1553. [PMID: 38672634 PMCID: PMC11049592 DOI: 10.3390/cancers16081553] [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: 03/12/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
There is growing recognition of early-onset gastrointestinal (GI) malignancies in young adults < 50 years of age. While much of the literature has emphasized colorectal cancer, these also include esophageal, gastric, liver, pancreatic, and biliary tract malignancies. Various factors, including lifestyle, hereditary, and environmental elements, have been proposed to explain the rising incidence of GI malignancies in the younger population. This review aims to provide an overview of the recent literature, including global trends and information regarding genetic and environmental risk factors.
Collapse
Affiliation(s)
- Aayush Vishwanath
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA;
| | - Shreyas Krishna
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (A.P.M.)
| | - Albert P. Manudhane
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (A.P.M.)
| | - Phil A. Hart
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (A.P.M.)
| | - Somashekar G. Krishna
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University, Columbus, OH 43210, USA; (S.K.); (A.P.M.)
| |
Collapse
|
3513
|
Gao P, Li X, He Z, Zhang H, Zhang Z, Liu Z. Lethal Immune Myocarditis and Myasthenia Gravis Due to Anti-PD-1 Treatment for a Bladder Cancer Patient: A Case Report and Possible Treatment Inspiration. Int Med Case Rep J 2024; 17:359-365. [PMID: 38651079 PMCID: PMC11034559 DOI: 10.2147/imcrj.s449525] [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: 12/05/2023] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) have become a new hope for many patients with advanced cancer by blocking tumor immune escape. Bladder cancer is a common malignant tumor of the urinary tract epithelium that often relapses and metastasizes after surgery, chemotherapy, and radiotherapy. Immunotherapy has dramatically improved patient survival rates and clinical benefits as a new, potentially effective therapy. However, avoidance of various immune-related adverse events (irAEs) remains an implausible idea. ICI-induced myocarditis is different from viral myocarditis, and mortality is still high with the current treatment. We report the case of an 82-year-old female patient with ICI-induced fulminant myocarditis and myasthenia gravis. Although she actively accepted the current mainstream treatment for immune-related myocarditis and myasthenia, she died of heart and respiratory failure. Analyzing and reporting the patient's disease development process and the changes in related indicators may help peers gain a deeper understanding of immune-related adverse events and reduce the mortality of immune-related myocarditis.
Collapse
Affiliation(s)
- Pan Gao
- Department of Urology, Second People’s Hospital of Yichang, Yichang, Hubei Province, People’s Republic of China
- Department of Urology, Second People’s Hospital of China Three Gorges University, Yichang, Hubei Province, People’s Republic of China
| | - Xinyu Li
- Department of Urology, Second People’s Hospital of Yichang, Yichang, Hubei Province, People’s Republic of China
- Department of Urology, Second People’s Hospital of China Three Gorges University, Yichang, Hubei Province, People’s Republic of China
| | - Ziqiu He
- Department of Urology, Second People’s Hospital of Yichang, Yichang, Hubei Province, People’s Republic of China
- Department of Urology, Second People’s Hospital of China Three Gorges University, Yichang, Hubei Province, People’s Republic of China
| | - Hongbo Zhang
- Department of Urology, Second People’s Hospital of Yichang, Yichang, Hubei Province, People’s Republic of China
- Department of Urology, Second People’s Hospital of China Three Gorges University, Yichang, Hubei Province, People’s Republic of China
| | - Zhi Zhang
- Department of Urology, Second People’s Hospital of Yichang, Yichang, Hubei Province, People’s Republic of China
- Department of Urology, Second People’s Hospital of China Three Gorges University, Yichang, Hubei Province, People’s Republic of China
| | - Zonglai Liu
- Department of Urology, Second People’s Hospital of Yichang, Yichang, Hubei Province, People’s Republic of China
- Department of Urology, Second People’s Hospital of China Three Gorges University, Yichang, Hubei Province, People’s Republic of China
| |
Collapse
|
3514
|
Cunningham ML, Schiewer MJ. PARP-ish: Gaps in Molecular Understanding and Clinical Trials Targeting PARP Exacerbate Racial Disparities in Prostate Cancer. Cancer Res 2024; 84:743102. [PMID: 38635890 PMCID: PMC11217733 DOI: 10.1158/0008-5472.can-23-3458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/25/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
PARP is a nuclear enzyme with a major function in the DNA damage response. PARP inhibitors (PARPi) have been developed for treating tumors harboring homologous recombination repair (HRR) defects that lead to a dependency on PARP. There are currently three PARPi approved for use in advanced prostate cancer (PCa), and several others are in clinical trials for this disease. Recent clinical trial results have reported differential efficacy based on the specific PARPi utilized as well as patient race. There is a racial disparity in PCa, where African American (AA) males are twice as likely to develop and die from the disease compared to European American (EA) males. Despite the disparity, there continues to be a lack of diversity in clinical trial cohorts for PCa. In this review, PARP nuclear functions, inhibition, and clinical relevance are explored through the lens of racial differences. This review will touch on the biological variations that have been explored thus far between AA and EA males with PCa to offer rationale for investigating PARPi response in the context of race at both the basic science and the clinical development levels.
Collapse
Affiliation(s)
- Moriah L. Cunningham
- Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
| | - Matthew J. Schiewer
- Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
3515
|
Dong Y, Huang H, Wang A. Effects of different exercise interventions on chemotherapy-related cognitive impairment in patients with breast cancer: a study protocol for systematic review and network meta-analysis. BMJ Open 2024; 14:e078934. [PMID: 38631832 PMCID: PMC11029385 DOI: 10.1136/bmjopen-2023-078934] [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: 08/16/2023] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
INTRODUCTION Breast cancer stands as the most prevalent type of cancer affecting women globally, and chemotherapy plays a pivotal role in its treatment by diminishing tumour recurrence and enhancing the survival rates of patients. However, chemotherapy-related cognitive impairment (CRCI) often occurs in patients undergoing treatment. Although multiple clinical trials have indicated that exercise therapy can improve CRCI in patients with breast cancer, there are variations in the types of exercise interventions and their effectiveness. We aim to perform a pioneering network meta-analysis (NMA) to assess and prioritise the effectiveness of various exercise interventions in enhancing cognitive function in patients with breast cancer undergoing chemotherapy. METHODS AND ANALYSIS We will search multiple databases, including PubMed, Web of Science, Cochrane, Embase, China National Knowledge Infrastructure, VIP Database for Chinese Technical Periodicals, Wanfang and Sinomed databases, from their inception to May 2023. The main outcome is the cognitive function changes in patients with breast cancer, including subjective and objective results. We will specifically include randomised controlled trials reported in English and Chinese languages, whose primary outcome consists of an assessment of the cognitive function of patients with breast cancer using standardised and validated assessment tools, encompassing both subjective and objective outcomes. The quality of all the trials included will be evaluated based on 'Version 2 of the Cochrane tool for assessing the risk of bias in randomized controlled trials (RoB2)'. We will conduct a Bayesian NMA to thoroughly evaluate and compare the effectiveness of different exercise interventions. We will use cumulative ranking probability plots to estimate the ranking of the best interventions for various exercises. Network plots and funnel plots will be employed to display the study sizes and participants of each exercise intervention, as well as potential publication biases. ETHICS AND DISSEMINATION The study findings will be shared via peer-reviewed journals to ensure the highest quality and credibility of the research. As the reporting will not include any private patient data, there are no ethical considerations associated with this protocol. PROSPERO REGISTRATION NUMBER CRD42023406597.
Collapse
Affiliation(s)
- Yu Dong
- The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hao Huang
- The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Aiping Wang
- The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
3516
|
Ji P, He J. Prognostic value of pretreatment systemic immune-inflammation index in patients with endometrial cancer: a meta-analysis. Biomark Med 2024; 18:345-356. [PMID: 38623927 PMCID: PMC11218804 DOI: 10.2217/bmm-2023-0629] [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: 10/07/2023] [Accepted: 03/06/2024] [Indexed: 04/17/2024] Open
Abstract
Background: The present work focused on evaluating the systemic immune-inflammation index (SII) for its role in predicting endometrial cancer (EC) patient prognosis by meta-analysis. Methods: SII's role in predicting the prognosis of EC patients was analyzed by calculating combined hazard ratios (HRs) and 95% CIs. Results: As revealed by combined analysis, an increased SII predicted poor overall survival (HR = 2.01; 95% CI = 1.58-2.57; p < 0.001) as well as inferior progression-free survival (HR = 1.87; 95% CI = 1.36-2.58; p < 0.001) of EC. Conclusion: An increased SII score significantly predicted poor overall survival and progression-free survival in subjects with EC. The SII is suitable for predicting short- and long-term prognoses of patients with EC.
Collapse
Affiliation(s)
- Pengtian Ji
- Department of Oncological Radiotherapy, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, Zhejiang, 313000, China
| | - Junjun He
- Clinical Laboratory, Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Huzhou, Zhejiang, China
| |
Collapse
|
3517
|
Wedig J, Jasani S, Mukherjee D, Lathrop H, Matreja P, Pfau T, D'Alesio L, Guenther A, Fenn L, Kaiser M, Torok MA, McGue J, Sizemore GM, Noonan AM, Dillhoff ME, Blaser BW, Frankel TL, Culp S, Hart PA, Cruz-Monserrate Z, Mace TA. CD200 is overexpressed in the pancreatic tumor microenvironment and predictive of overall survival. Cancer Immunol Immunother 2024; 73:96. [PMID: 38619621 PMCID: PMC11018596 DOI: 10.1007/s00262-024-03678-6] [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: 02/19/2024] [Accepted: 03/15/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic cancer is an aggressive disease with a 5 year survival rate of 13%. This poor survival is attributed, in part, to limited and ineffective treatments for patients with metastatic disease, highlighting a need to identify molecular drivers of pancreatic cancer to target for more effective treatment. CD200 is a glycoprotein that interacts with the receptor CD200R and elicits an immunosuppressive response. Overexpression of CD200 has been associated with differential outcomes, depending on the tumor type. In the context of pancreatic cancer, we have previously reported that CD200 is expressed in the pancreatic tumor microenvironment (TME), and that targeting CD200 in murine tumor models reduces tumor burden. We hypothesized that CD200 is overexpressed on tumor and stromal populations in the pancreatic TME and that circulating levels of soluble CD200 (sCD200) have prognostic value for overall survival. We discovered that CD200 was overexpressed on immune, stromal, and tumor populations in the pancreatic TME. Particularly, single-cell RNA-sequencing indicated that CD200 was upregulated on inflammatory cancer-associated fibroblasts. Cytometry by time of flight analysis of PBMCs indicated that CD200 was overexpressed on innate immune populations, including monocytes, dendritic cells, and monocytic myeloid-derived suppressor cells. High sCD200 levels in plasma correlated with significantly worse overall and progression-free survival. Additionally, sCD200 correlated with the ratio of circulating matrix metalloproteinase (MMP) 3: tissue inhibitor of metalloproteinase (TIMP) 3 and MMP11/TIMP3. This study highlights the importance of CD200 expression in pancreatic cancer and provides the rationale for designing novel therapeutic strategies that target this protein.
Collapse
Affiliation(s)
- Jessica Wedig
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Molecular, Cellular and Developmental Biology Program, The Ohio State University, Columbus, USA
| | - Shrina Jasani
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Debasmita Mukherjee
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Molecular, Cellular and Developmental Biology Program, The Ohio State University, Columbus, USA
| | - Hannah Lathrop
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Priya Matreja
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Timothy Pfau
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Liliana D'Alesio
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Abigail Guenther
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Lexie Fenn
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Morgan Kaiser
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Molly A Torok
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
| | - Jake McGue
- Department of Surgical Oncology, University of Michigan, Ann Arbor, USA
| | - Gina M Sizemore
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Radiation Oncology, The Ohio State University, Columbus, USA
| | - Anne M Noonan
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Mary E Dillhoff
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Internal Medicine, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Bradley W Blaser
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Internal Medicine, Division of Hematology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Timothy L Frankel
- Department of Surgical Oncology, University of Michigan, Ann Arbor, USA
| | - Stacey Culp
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, USA
| | - Phil A Hart
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, 420 W. 12th Ave., Columbus, OH, 43210, USA
| | - Zobeida Cruz-Monserrate
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, 420 W. 12th Ave., Columbus, OH, 43210, USA
| | - Thomas A Mace
- The James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, USA.
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, 420 W. 12th Ave., Columbus, OH, 43210, USA.
| |
Collapse
|
3518
|
Maleki Z, Hassanzadeh J, Ghaem H. Correlation between socioeconomic indices and epidemiological indices of thyroid cancer from 1990 to 2019 year: a global ecologic study. BMC Cancer 2024; 24:467. [PMID: 38622568 PMCID: PMC11017482 DOI: 10.1186/s12885-024-12176-y] [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: 02/04/2024] [Accepted: 03/24/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND The incidence of thyroid cancer as the most common type of endocrine gland malignancy has risen more significantly than any malignancies in recent years. Estimated new cases of thyroid cancer in the United States in 2024 were 12,500 and 31,520 for men and women, respectively, and estimated deaths were 1,180 for women and 990 for men. Indices of socio-economic have been commonly used to measure the development of countries. Therefore, this study aimed to examine the correlation between indices of socioeconomic status and epidemiological indices of thyroid cancer throughout the world. In addition, this study has compared two indices of human development and a socio-demographic index. METHOD This worldwide ecological study used data on thyroid cancer incidence, mortality, human development index (HDI), and sociodemographic index (SDI) between 1990 and 2019 from the Global Burden of Disease (GBD). We evaluated the correlation between incidence and mortality rates with socioeconomic indices by using Pearson's correlation coefficient. Furthermore, for the first time, the generalized additive model (GAM) was employed for modeling. The statistical software R, version 4.2.2, was used to conduct all statistical analyses. RESULTS The correlation between the incidence of thyroid cancer and the HDI was significant and positive (r = 0.47, p-value < 0.001). While the correlation between thyroid cancer mortality and HDI was not statistically significant (r = 0.01, p-value = 0.076). Besides, the incidence of thyroid cancer was significantly positively correlated with SDI (r = 0.48, p-value < 0.001). The multiple GAM showed that for one unit increase in HDI, the risk of thyroid cancer was increased by 2.1 times (RR = 2.1, 95%CI = 2.04 to 2.19), and for one unit increase in SDI, the risk of thyroid cancer was shown to increase by 2.2 times. (RR = 2.2, 95%CI = 2.19 to 2.35). CONCLUSION It has been evident that countries with higher incidence of thyroid cancer display higher socioeconomic indices. While, countries with higher socioeconomic indices, report lower mortality rates. However, based on the modeling results, it can be concluded that the SDI is slightly more useful in this regard. Therefore, examining the epidemiological indices of thyroid cancer by socio-economic indices can be useful to reflect a clear image of the distribution of this cancer in each country, and can be used for planning cancer prevention strategies.
Collapse
Affiliation(s)
- Zahra Maleki
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Hassanzadeh
- Department of Epidemiology, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Haleh Ghaem
- Non-Communicable Diseases Research Center, Department of Epidemiology, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3519
|
Kim B, Jung J. Metabolomic Approach to Identify Potential Biomarkers in KRAS-Mutant Pancreatic Cancer Cells. Biomedicines 2024; 12:865. [PMID: 38672219 PMCID: PMC11048406 DOI: 10.3390/biomedicines12040865] [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: 03/28/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Pancreatic cancer is characterized by its high mortality rate and limited treatment options, often driven by oncogenic RAS mutations. In this study, we investigated the metabolomic profiles of pancreatic cancer cells based on their KRAS genetic status. Utilizing both KRAS-wildtype BxPC3 and KRAS-mutant PANC1 cell lines, we identified 195 metabolites differentially altered by KRAS status through untargeted metabolomics. Principal component analysis and hierarchical condition trees revealed distinct separation between KRAS-wildtype and KRAS-mutant cells. Metabolite set enrichment analysis highlighted significant pathways such as homocysteine degradation and taurine and hypotaurine metabolism. Additionally, lipid enrichment analysis identified pathways including fatty acyl glycosides and sphingoid bases. Mapping of identified metabolites to KEGG pathways identified nine significant metabolic pathways associated with KRAS status, indicating diverse metabolic alterations in pancreatic cancer cells. Furthermore, we explored the impact of TRPML1 inhibition on the metabolomic profile of KRAS-mutant pancreatic cancer cells. TRPML1 inhibition using ML-SI1 significantly altered the metabolomic profile, leading to distinct separation between vehicle-treated and ML-SI1-treated PANC1 cells. Metabolite set enrichment analysis revealed enriched pathways such as arginine and proline metabolism, and mapping to KEGG pathways identified 17 significant metabolic pathways associated with TRPML1 inhibition. Interestingly, some metabolites identified in PANC1 compared to BxPC3 were oppositely regulated by TRPML1 inhibition, suggesting their potential as biomarkers for KRAS-mutant cancer cells. Overall, our findings shed light on the distinct metabolite changes induced by both KRAS status and TRPML1 inhibition in pancreatic cancer cells, providing insights into potential therapeutic targets and biomarkers for this deadly disease.
Collapse
Affiliation(s)
| | - Jewon Jung
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan 48434, Republic of Korea;
| |
Collapse
|
3520
|
Muaddi H, Kearse L, Warner S. Multimodal Approaches to Patient Selection for Pancreas Cancer Surgery. Curr Oncol 2024; 31:2260-2273. [PMID: 38668070 PMCID: PMC11049254 DOI: 10.3390/curroncol31040167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
With an overall 5-year survival rate of 12%, pancreas ductal adenocarcinoma (PDAC) is an aggressive cancer that claims more than 50,000 patient lives each year in the United States alone. Even those few patients who undergo curative-intent resection with favorable pathology reports are likely to experience recurrence within the first two years after surgery and ultimately die from their cancer. We hypothesize that risk factors for these early recurrences can be identified with thorough preoperative staging, thus enabling proper patient selection for surgical resection and avoiding unnecessary harm. Herein, we review evidence supporting multidisciplinary and multimodality staging, comprehensive neoadjuvant treatment strategies, and optimal patient selection for curative-intent surgical resections. We further review data generated from our standardized approach at the Mayo Clinic and extrapolate to inform potential future investigations.
Collapse
Affiliation(s)
| | | | - Susanne Warner
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN 55902, USA; (H.M.)
| |
Collapse
|
3521
|
Zeng Z, Zhu J, Wang Z, Wang G, Yan J, Zhang F. Pelvic target volume inter-fractional motion during radiotherapy for cervical cancer with daily iterative cone beam computed tomography. Radiat Oncol 2024; 19:48. [PMID: 38622628 PMCID: PMC11017626 DOI: 10.1186/s13014-024-02438-1] [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: 01/08/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Tumor regression and organ movements indicate that a large margin is used to ensure target volume coverage during radiotherapy. This study aimed to quantify inter-fractional movements of the uterus and cervix in patients with cervical cancer undergoing radiotherapy and to evaluate the clinical target volume (CTV) coverage. METHODS This study analyzed 303 iterative cone beam computed tomography (iCBCT) scans from 15 cervical cancer patients undergoing external beam radiotherapy. CTVs of the uterus (CTV-U) and cervix (CTV-C) contours were delineated based on each iCBCT image. CTV-U encompassed the uterus, while CTV-C included the cervix, vagina, and adjacent parametrial regions. Compared with the planning CTV, the movement of CTV-U and CTV-C in the anterior-posterior, superior-inferior, and lateral directions between iCBCT scans was measured. Uniform expansions were applied to the planning CTV to assess target coverage. RESULTS The motion (mean ± standard deviation) in the CTV-U position was 8.3 ± 4.1 mm in the left, 9.8 ± 4.4 mm in the right, 12.6 ± 4.0 mm in the anterior, 8.8 ± 5.1 mm in the posterior, 5.7 ± 5.4 mm in the superior, and 3.0 ± 3.2 mm in the inferior direction. The mean CTV-C displacement was 7.3 ± 3.2 mm in the left, 8.6 ± 3.8 mm in the right, 9.0 ± 6.1 mm in the anterior, 8.4 ± 3.6 mm in the posterior, 5.0 ± 5.0 mm in the superior, and 3.0 ± 2.5 mm in the inferior direction. Compared with the other tumor (T) stages, CTV-U and CTV-C motion in stage T1 was larger. A uniform CTV planning treatment volume margin of 15 mm failed to encompass the CTV-U and CTV-C in 11.1% and 2.2% of all fractions, respectively. The mean volume change of CTV-U and CTV-C were 150% and 51%, respectively, compared with the planning CTV. CONCLUSIONS Movements of the uterine corpus are larger than those of the cervix. The likelihood of missing the CTV is significantly increased due to inter-fractional motion when utilizing traditional planning margins. Early T stage may require larger margins. Personal radiotherapy margining is needed to improve treatment accuracy.
Collapse
Affiliation(s)
- Zheng Zeng
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 100730, Beijing, China
| | - Jiawei Zhu
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 100730, Beijing, China
| | - Zhiqun Wang
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 100730, Beijing, China
| | - Guangyu Wang
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 100730, Beijing, China
| | - Junfang Yan
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 100730, Beijing, China.
| | - Fuquan Zhang
- Department of Radiation Oncology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 100730, Beijing, China.
| |
Collapse
|
3522
|
Ben-Eltriki M, Gayle EJ, Paras JM, Nyame-Addo L, Chhabra M, Deb S. Vitamin D in Melanoma: Potential Role of Cytochrome P450 Enzymes. Life (Basel) 2024; 14:510. [PMID: 38672780 PMCID: PMC11050855 DOI: 10.3390/life14040510] [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: 02/25/2024] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Vitamin D is a promising anticancer agent for the prevention and treatment of several cancers, including melanoma. Low 25-hydroxyvitamin D levels, a routinely used marker for vitamin D, have been suggested as one of the factors in the development and progression of melanoma. The parent vitamin D needs activation by cytochrome P450 (CYP) enzymes to exert its actions via the vitamin D receptor (VDR). This review discusses the role of vitamin D in melanoma and how CYP-mediated metabolism can potentially affect the actions of vitamin D. Through interacting with the retinoid X receptor, VDR signaling leads to anti-inflammatory, antioxidative, and anticancer actions. Calcitriol, the dihydroxylated form of vitamin D3, is the most active and potent ligand of VDR. CYP27A1, CYP27B1, and CYP2R1 are involved in the activation of vitamin D, whereas CYP24A1 and CYP3A4 are responsible for the degradation of the active vitamin D. CYP24A1, the primary catabolic enzyme of calcitriol, is overexpressed in melanoma tissues and cells. Several drug classes and natural health products can modulate vitamin D-related CYP enzymes and eventually cause lower levels of vitamin D and its active metabolites in tissues. Although the role of vitamin D in the development of melanoma is yet to be fully elucidated, it has been proposed that melanoma prevention may be significantly aided by increased vitamin D signaling. Furthermore, selective targeting of the catabolic enzymes responsible for vitamin D degradation could be a plausible strategy in melanoma therapy. Vitamin D signaling can be improved by utilizing dietary supplements or by modulating CYP metabolism. A positive association exists between the intake of vitamin D supplements and improved prognosis for melanoma patients. Further investigation is required to determine the function of vitamin D supplementation and specific enzyme targeting in the prevention of melanoma.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- Clinical Pharmacology Lab, Department of Pharmacology and Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
- Cochrane Hypertension Review Group, Therapeutic Initiative, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Erysa J. Gayle
- College of Biomedical Sciences, Larkin University, Miami, FL 33169, USA; (E.J.G.); (J.M.P.)
| | - Jhoanne M. Paras
- College of Biomedical Sciences, Larkin University, Miami, FL 33169, USA; (E.J.G.); (J.M.P.)
| | - Louisa Nyame-Addo
- College of Biomedical Sciences, Larkin University, Miami, FL 33169, USA; (E.J.G.); (J.M.P.)
| | - Manik Chhabra
- Clinical Pharmacology Lab, Department of Pharmacology and Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| | - Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA
| |
Collapse
|
3523
|
Wang J, Cai L, Huang G, Wang C, Zhang Z, Xu J. CENPA and BRCA1 are potential biomarkers associated with immune infiltration in heart failure and pan-cancer. Heliyon 2024; 10:e28786. [PMID: 38576566 PMCID: PMC10990859 DOI: 10.1016/j.heliyon.2024.e28786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
Heart failure (HF) and cancer are the two leading causes of death worldwide and affect one another in a bidirectional way. We aimed to identify hub therapeutic genes as potential biomarkers for the identification and treatment of HF and cancer. Gene expression data of heart samples from patients with ischemic HF (IHF) and healthy controls were retrieved from the GSE42955 and GSE57338 databases. Difference analysis and weighted gene co-expression network analysis (WGCNA) were used to identify key modules associated with IHF. The overlapping genes were subjected to gene and protein enrichment analyses to construct a protein-protein interaction (PPI) network, which was screened for hub genes among the overlapping genes. A total of eight hub genes were subjected to correlation, immune cell infiltration, and ROC analyses. Then we analyzed the roles of two significant genes in 33 tumor types to explore their potential as common targets in HF and cancer. A total of 85 genes were identified by WGCNA and differentially expressed gene (DEG) analyses. BRCA1, MED17, CENPA, RXRA, RXRB, SMARCA2, CDCA2, and PMS2 were identified as the hub genes with IHF. Finally, CENPA and BRCA1 were identified as potential common targets for IHF and cancer. These findings provide new perspectives for expanding our understanding of the etiology and underlying mechanisms of HF and cancer.
Collapse
Affiliation(s)
- Jian Wang
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, 82 Qinglong Street, Chengdu, 610014, China
- Department of Cardiology, The Third People's Hospital of Chengdu, 82 Qinglong Street, Chengdu, 610014, China
| | - Lin Cai
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, 82 Qinglong Street, Chengdu, 610014, China
- Department of Cardiology, The Third People's Hospital of Chengdu, 82 Qinglong Street, Chengdu, 610014, China
| | - Gang Huang
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, 82 Qinglong Street, Chengdu, 610014, China
- Department of Cardiology, The Third People's Hospital of Chengdu, 82 Qinglong Street, Chengdu, 610014, China
| | - Chunbin Wang
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, 82 Qinglong Street, Chengdu, 610014, China
- Department of Cardiology, The Third People's Hospital of Chengdu, 82 Qinglong Street, Chengdu, 610014, China
| | - Zhen Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, 82 Qinglong Street, Chengdu, 610014, China
- Department of Cardiology, The Third People's Hospital of Chengdu, 82 Qinglong Street, Chengdu, 610014, China
- Chengdu Institute of Cardiovascular Disease, 82 Qinglong Street, Chengdu, 610014, China
| | - Junbo Xu
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, 82 Qinglong Street, Chengdu, 610014, China
- Department of Cardiology, The Third People's Hospital of Chengdu, 82 Qinglong Street, Chengdu, 610014, China
- Chengdu Institute of Cardiovascular Disease, 82 Qinglong Street, Chengdu, 610014, China
| |
Collapse
|
3524
|
Iqbal S, Karim MR, Mohammad S, Ahn JC, Kariyarath Valappil A, Mathiyalagan R, Yang DC, Jung DH, Bae H, Yang DU. In Silico and In Vitro Study of Isoquercitrin against Kidney Cancer and Inflammation by Triggering Potential Gene Targets. Curr Issues Mol Biol 2024; 46:3328-3341. [PMID: 38666938 PMCID: PMC11049307 DOI: 10.3390/cimb46040208] [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: 03/05/2024] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Kidney cancer has emerged as a major medical problem in recent times. Multiple compounds are used to treat kidney cancer by triggering cancer-causing gene targets. For instance, isoquercitrin (quercetin-3-O-β-d-glucopyranoside) is frequently present in fruits, vegetables, medicinal herbs, and foods and drinks made from plants. Our previous study predicted using protein-protein interaction (PPI) and molecular docking analysis that the isoquercitrin compound can control kidney cancer and inflammation by triggering potential gene targets of IGF1R, PIK3CA, IL6, and PTGS2. So, the present study is about further in silico and in vitro validation. We performed molecular dynamic (MD) simulation, gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, cytotoxicity assay, and RT-PCR and qRT-PCR validation. According to the MD simulation (250 ns), we found that IGF1R, PIK3CA, and PTGS2, except for IL6 gene targets, show stable binding energy with a stable complex with isoquercitrin. We also performed gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of the final targets to determine their regulatory functions and signaling pathways. Furthermore, we checked the cytotoxicity effect of isoquercitrin (IQ) and found that 5 μg/mL and 10 μg/mL doses showed higher cell viability in a normal kidney cell line (HEK 293) and also inversely showed an inhibition of cell growth at 35% and 45%, respectively, in the kidney cancer cell line (A498). Lastly, the RT-PCR and qRT-PCR findings showed a significant decrease in PTGS2, PIK3CA, and IGF1R gene expression, except for IL6 expression, following dose-dependent treatments with IQ. Thus, we can conclude that isoquercitrin inhibits the expression of PTGS2, PIK3CA, and IGF1R gene targets, which in turn controls kidney cancer and inflammation.
Collapse
Affiliation(s)
- Safia Iqbal
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.I.); (M.R.K.); (A.K.V.); (D.-C.Y.)
| | - Md. Rezaul Karim
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.I.); (M.R.K.); (A.K.V.); (D.-C.Y.)
| | - Shahnawaz Mohammad
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.M.); (J.C.A.); (R.M.); (D.-H.J.)
| | - Jong Chan Ahn
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.M.); (J.C.A.); (R.M.); (D.-H.J.)
| | - Anjali Kariyarath Valappil
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.I.); (M.R.K.); (A.K.V.); (D.-C.Y.)
| | - Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.M.); (J.C.A.); (R.M.); (D.-H.J.)
| | - Deok-Chun Yang
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.I.); (M.R.K.); (A.K.V.); (D.-C.Y.)
| | - Dae-Hyo Jung
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.M.); (J.C.A.); (R.M.); (D.-H.J.)
| | - Hyocheol Bae
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Gyeonggi-do, Republic of Korea
| | - Dong Uk Yang
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea; (S.I.); (M.R.K.); (A.K.V.); (D.-C.Y.)
| |
Collapse
|
3525
|
Pothuraju R, Khan I, Jain M, Bouvet M, Malafa M, Roy HK, Kumar S, Batra SK. Colorectal cancer murine models: Initiation to metastasis. Cancer Lett 2024; 587:216704. [PMID: 38360138 PMCID: PMC11257378 DOI: 10.1016/j.canlet.2024.216704] [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: 10/23/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/17/2024]
Abstract
Despite significant advancements in prevention and treatment, colorectal cancer (CRC) remains the third leading cause of cancer-related deaths. Animal models, including xenografts, syngeneic, and genetically engineered, have emerged as indispensable tools in cancer research. These models offer a valuable platform to address critical questions regarding molecular pathogenesis and test therapeutic interventions before moving on to clinical trials. Advancements in CRC animal models have also facilitated the advent of personalized and precision medicine. Patient-derived xenografts and genetically engineered mice that mirror features of human tumors allow for tailoring treatments to specific CRC subtypes, improving treatment outcomes and quality of life. To overcome the limitations of individual model systems, recent studies have employed a multi-modal approach, combining different animal models, 3D organoids, and in vitro studies. This integrative approach provides a comprehensive understanding of CRC biology, including the tumor microenvironment and therapeutic responses, driving the development of more effective and personalized therapeutic interventions. This review discusses the animal models used for CRC research, including recent advancements and limitations of these animal models.
Collapse
Affiliation(s)
- Ramesh Pothuraju
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA; Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India
| | - Imran Khan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, California, USA
| | - Mokenge Malafa
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Hemant K Roy
- Department of Medicine, Baylor College of Medicine, Houston, TX-77030, USA
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE-68198, USA.
| |
Collapse
|
3526
|
Awais M, Ahmad R, Kausar N, Alzahrani AI, Alalwan N, Masood A. ALL classification using neural ensemble and memetic deep feature optimization. Front Artif Intell 2024; 7:1351942. [PMID: 38655268 PMCID: PMC11035867 DOI: 10.3389/frai.2024.1351942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/15/2024] [Indexed: 04/26/2024] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a fatal blood disorder characterized by the excessive proliferation of immature white blood cells, originating in the bone marrow. An effective prognosis and treatment of ALL calls for its accurate and timely detection. Deep convolutional neural networks (CNNs) have shown promising results in digital pathology. However, they face challenges in classifying different subtypes of leukemia due to their subtle morphological differences. This study proposes an improved pipeline for binary detection and sub-type classification of ALL from blood smear images. At first, a customized, 88 layers deep CNN is proposed and trained using transfer learning along with GoogleNet CNN to create an ensemble of features. Furthermore, this study models the feature selection problem as a combinatorial optimization problem and proposes a memetic version of binary whale optimization algorithm, incorporating Differential Evolution-based local search method to enhance the exploration and exploitation of feature search space. The proposed approach is validated using publicly available standard datasets containing peripheral blood smear images of various classes of ALL. An overall best average accuracy of 99.15% is achieved for binary classification of ALL with an 85% decrease in the feature vector, together with 99% precision and 98.8% sensitivity. For B-ALL sub-type classification, the best accuracy of 98.69% is attained with 98.7% precision and 99.57% specificity. The proposed methodology shows better performance metrics as compared with several existing studies.
Collapse
Affiliation(s)
- Muhammad Awais
- Department of Electrical and Computer Engineering, COMSATS University Islamabad, Wah, Pakistan
- Department of Computer Engineering, TED University Ankara, Ankara, Türkiye
| | - Riaz Ahmad
- Department of Computer Science, Iqra University Islamabad, Islamabad, Pakistan
- Department of Computer Science, COMSATS University Islamabad, Wah, Pakistan
| | - Nabeela Kausar
- Department of Computer Science, Iqra University Islamabad, Islamabad, Pakistan
| | - Ahmed Ibrahim Alzahrani
- Department of Computer Science, Community College, King Saud University, Riyadh, Saudi Arabia
| | - Nasser Alalwan
- Department of Computer Science, Community College, King Saud University, Riyadh, Saudi Arabia
| | - Anum Masood
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Radiology, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
3527
|
Galal ER, Abdelhakam DA, Ahmed LK, Elhusseny Y, Sayed SEP, Eltaweel NH. The association of FSCN1 (rs852479, rs1640233) and HOTAIR (rs920778) polymorphisms with the risk of breast cancer in Egyptian women. Mol Biol Rep 2024; 51:495. [PMID: 38587571 PMCID: PMC11001669 DOI: 10.1007/s11033-024-09459-9] [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: 02/22/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Breast cancer (BC) is one of the most prevalent cancers that contribute to mortality among women worldwide. Despite contradictory findings, considerable evidence suggests that single nucleotide polymorphisms (SNPs) in the FSCN1 and HOTAIR genes may have a causative impact on the development of BC. This case-control study was conducted to evaluate the association of genotype frequency in FSCN1 rs852479, rs1640233, and HOTAIR rs920778 with susceptibility and prognosis of BC, as well as the impact of clinical stages and hormonal features. METHODS AND RESULTS FSCN1 (rs852479, rs1640233) and HOTAIR (rs920778) were genotyped using TaqMan real-time PCR assay in 200 BC patients and 200 cancer-free controls, all representing Egyptian women. Genotypic analyses in association with clinicopathological factors and disease risk were assessed. As a result, a significant association with BC risk was observed for CC genotype frequency of FSCN1 rs852479 A > C (OR = 0.395, 95% CI 0.204-0.76, p-value = 0.005). However, no significant correlation was detected between the FSCN1 rs1640233 C > T and HOTAIR rs920778 C > T polymorphic variants and susceptibility to BC. Interestingly, CC genotype of FSCN1 rs1640233 was more likely to progress tumor size and lymph node invasion in BC cases (p-value = 0.04 and 0.02, respectively). Moreover, it was revealed that there was a non-significant correlation between the haplotype distributions of FSCN1 rs852479 and rs1640233 and the probability of BC. CONCLUSIONS Based on the sample size and genetic characteristics of the subjects involved in the present study, our findings indicated that FSCN1 rs852479 may contribute to BC susceptibility in a sample of the Egyptian population.
Collapse
Affiliation(s)
- Eman Reda Galal
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt.
| | - Dina A Abdelhakam
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Lamiaa Khalaf Ahmed
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Yasmine Elhusseny
- Medical Biochemistry and Molecular Biology Department, School of Medicine, Newgiza University, Giza, Egypt
| | - Sherif El Prince Sayed
- Department of General Surgery, Faculty of Medicine, Beni-Suef University, Beni Suef, Egypt
| | - Noha H Eltaweel
- Medical Molecular Genetics Department, Human Genetics and Genome Project Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
3528
|
Wang Y, Li Y, Jing Y, Yang Y, Wang H, Ismtula D, Guo C. Tubulin alpha-1b chain was identified as a prognosis and immune biomarker in pan-cancer combing with experimental validation in breast cancer. Sci Rep 2024; 14:8201. [PMID: 38589634 PMCID: PMC11001892 DOI: 10.1038/s41598-024-58982-z] [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: 01/18/2024] [Accepted: 04/05/2024] [Indexed: 04/10/2024] Open
Abstract
The α-tubulin subtype, Tubulin α-1b chain (TUBA1B), has been shown to influence immune cell infiltration, cancer growth, and survival across various malignancies. However, a comprehensive study has not yet been undertaken examining the immunological and predictive effects of TUBA1B in a pan-carcinoma context. Using data from TCGA, GEO, and other databases, we analyzed TUBA1B expression across various carcinoma types using transcriptional profiling, prognostic implications, genetic and epigenetic alterations, methylation patterns, and immunological significance. To validate our findings, we conducted Western blot analysis to assess TUBA1B protein levels in matched breast cancer tissue samples and performed CCK-8 proliferation assay, flow cytometry, transwell invasion, and migration assays to comprehensively examine the functional impact of TUBA1B on breast cancer cells. Our pan-cancer analysis found TUBA1B upregulation across most tumor types, with varying expression patterns in distinct immune and molecular subtypes. High TUBA1B expression was an independent risk factor and associated with poor prognoses in several cancers, including BRCA, KICH, LGG, LUAD, and MESO. TUBA1B also demonstrates moderate to high diagnostic accuracy in most tumor types. Increased m6A methylation levels were observed in the TUBA1B gene, while its promoter region displayed low methylation levels. TUBA1B's expression impacted some cancers by elevating tumor mutation burden, microsatellite instability, neoantigen formation, immune cell infiltration, and the modulation of immune checkpoints. Functional enrichment analysis highlights TUBA1B's involvement in important cellular processes such as the cell cycle, p53 signaling, cell senescence, programmed cell death, and the regulation of immune-related pathways. Moreover, our study reveals higher TUBA1B protein expression in breast cancer tissues compared to adjacent tissues. In vitro experiments confirm that TUBA1B deletion reduces breast cancer cell proliferation, invasion, and migration while increasing apoptosis. In conclusion, our study suggests that TUBA1B could potentially serve as a diagnostic marker for predicting cancer immunological profiles and survival outcomes and shed light on the expression and role of TUBA1B in breast cancer, providing a solid foundation for considering it as a promising therapeutic target for breast cancer patient treatment.
Collapse
Affiliation(s)
- Yiyang Wang
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yongxiang Li
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yubo Jing
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yuqi Yang
- The First Clinical Medical College of Xinjiang Medical University, Urumqi, 830054, China
| | - Haiyan Wang
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Dilimulati Ismtula
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Chenming Guo
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
| |
Collapse
|
3529
|
Lang J, McClure TD, Margolis DJA. MRI-Ultrasound Fused Approach for Prostate Biopsy-How It Is Performed. Cancers (Basel) 2024; 16:1424. [PMID: 38611102 PMCID: PMC11010881 DOI: 10.3390/cancers16071424] [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: 02/27/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
The use of MRI-ultrasound image fusion targeted biopsy of the prostate in the face of an elevated serum PSA is now recommended by multiple societies, and results in improved detection of clinically significant cancer and, potentially, decreased detection of indolent disease. This combines the excellent sensitivity of MRI for clinically significant prostate cancer and the real-time biopsy guidance and confirmation of ultrasound. Both transperineal and transrectal approaches can be implemented using cognitive fusion, mechanical fusion with an articulated arm and electromagnetic registration, or pure software registration. The performance has been shown comparable to in-bore MRI biopsy performance. However, a number of factors influence the performance of this technique, including the quality and interpretation of the MRI, the approach used for biopsy, and experience of the practitioner, with most studies showing comparable performance of MRI-ultrasound fusion to in-bore targeted biopsy. Future improvements including artificial intelligence promise to refine the performance of all approaches.
Collapse
Affiliation(s)
- Jacob Lang
- Department of Urology, Weill Cornell Medicine, New York, NY 10068, USA
| | - Timothy Dale McClure
- Department of Urology, Weill Cornell Medicine, New York, NY 10068, USA
- Department of Radiology, Weill Cornell Medicine, New York, NY 10068, USA
| | | |
Collapse
|
3530
|
Barrientos-Toro EN, Ding Q, Raso MG. Translational Aspects in Metaplastic Breast Carcinoma. Cancers (Basel) 2024; 16:1433. [PMID: 38611109 PMCID: PMC11011105 DOI: 10.3390/cancers16071433] [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: 02/06/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Breast cancer is the most common cancer among women. Metaplastic breast carcinoma (MpBC) is a rare, heterogeneous group of invasive breast carcinomas, which are classified as predominantly triple-negative breast carcinomas (TNBCs; HR-negative/HER2-negative). Histologically, MpBC is classified into six subtypes. Two of these are considered low-grade and the others are high-grade. MpBCs seem to be more aggressive, less responsive to neoadjuvant chemotherapy, and have higher rates of chemoresistance than other TNBCs. MpBCs have a lower survival rate than expected for TNBCs. MpBC treatment represents a challenge, leading to a thorough exploration of the tumor immune microenvironment, which has recently opened the possibility of new therapeutic strategies. The epithelial-mesenchymal transition in MpBC is characterized by the loss of intercellular adhesion, downregulation of epithelial markers, underexpression of genes with biological epithelial functions, upregulation of mesenchymal markers, overexpression of genes with biological mesenchymal functions, acquisition of fibroblast-like (spindle) morphology, cytoskeleton reorganization, increased motility, invasiveness, and metastatic capabilities. This article reviews and summarizes the current knowledge and translational aspects of MpBC.
Collapse
Affiliation(s)
- Elizve Nairoby Barrientos-Toro
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Qingqing Ding
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| |
Collapse
|
3531
|
Koukourakis IM, Xanthopoulou E, Koukourakis MI, Tiniakos D, Kouloulias V, Zygogianni A. IFN-Type-I Response and Systemic Immunity in Rectal Adenocarcinoma Patients Treated with Conventional or Hypofractionated Neoadjuvant Radiotherapy. Biomolecules 2024; 14:448. [PMID: 38672465 PMCID: PMC11048635 DOI: 10.3390/biom14040448] [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: 02/28/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
The IFN-type-I pathway is involved in radiotherapy (RT)-mediated immune responses. Large RT fractions have been suggested to potently induce this pathway. Neoadjuvant hypofractionated short-course (scRT) and conventional long-course (lcRT) RT applied for the treatment of locally advanced rectal adenocarcinoma patients provides a unique model to address the immuno-stimulatory properties of RT on a systemic level. We prospectively analyzed the IFNβ plasma levels and lymphocyte counts (LCs) of rectal adenocarcinoma patients before and after treatment with scRT (n = 22) and lcRT (n = 40). Flow cytometry was conducted to assess the effects on lymphocytic subpopulations in a subset of 20 patients. A statistically significant increase in the post-RT IFNβ plasma levels was noted in patients undergoing scRT (p = 0.004). Improved pathological tumor regression was associated with elevated post-RT IFNβ levels (p = 0.003). Although all patients experienced substantial lymphopenia after treatment, the post-RT LC of patients treated with scRT were significantly higher compared to lcRT (p = 0.001). Patients undergoing scRT displayed significantly lower percentages of regulatory CD4+/CD25+ T-cells after therapy (p = 0.02). scRT enables effective stimulation of the IFN-type-I pathway on a systemic level and confers decreased lymphocytic cytotoxicity and limited regulatory T-cell activation compared to lcRT, supporting its increasing role in immuno-RT trials.
Collapse
Affiliation(s)
- Ioannis M. Koukourakis
- Radiation Oncology Unit, 1st Department of Radiology, School of Medicine, Aretaieion Hospital, National and Kapodistrian University of Athens (NKUOA), 11528 Athens, Greece;
| | - Erasmia Xanthopoulou
- Department of Radiotherapy/Oncology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.X.); (M.I.K.)
| | - Michael I. Koukourakis
- Department of Radiotherapy/Oncology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.X.); (M.I.K.)
| | - Dina Tiniakos
- Department of Pathology, Aretaieion University Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Vassilis Kouloulias
- Radiotherapy Unit, 2nd Department of Radiology, Attikon Hospital, School of Medicine, Rimini 1, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Anna Zygogianni
- Radiation Oncology Unit, 1st Department of Radiology, School of Medicine, Aretaieion Hospital, National and Kapodistrian University of Athens (NKUOA), 11528 Athens, Greece;
| |
Collapse
|
3532
|
Bezerra P, Motti EF. 3-NAntC: A Potent Crotoxin B-Derived Peptide against the Triple-Negative MDA-MB-231 Breast Cancer Cell Line. Molecules 2024; 29:1646. [PMID: 38611925 PMCID: PMC11013444 DOI: 10.3390/molecules29071646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/25/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Breast cancer stands as the most prevalent type of tumor and a significant contributor to cancer-related deaths. Among its various subtypes, triple-negative breast cancer (TNBC) presents the worst prognosis due to its aggressive nature and the absence of effective treatments. Crotoxin, a protein found in the venom of Crotalus genus snakes, has demonstrated notable antitumor activity against aggressive solid tumors. However, its application has been hindered by substantial toxicity in humans. In efforts to address this challenge, Crotoxin B-derived peptides were synthesized and evaluated in vitro for their antitumor potential, leading to the discovery of 3-NAntC. Treatment with 3-NAntC at 1 µg/mL for 72 h notably reduced the viability of MDA-MB-231 cells to 49.0 ± 17.5% (p < 0.0001), while exhibiting minimal impact on the viability of HMEC cells (98.2 ± 13.8%) under the same conditions. Notably, 3-NAntC displayed superior antitumoral activity in vitro compared to cisplatin and exhibited a similar effect to doxorubicin. Further investigation revealed that 3-NAntC decreased the proliferation of MDA-MB-231 cells and induced G2/M phase arrest. It primarily prompted optimal cell death by apoptosis, with a lower incidence of the less desirable cell death by necrosis in comparison to doxorubicin. Additionally, 3-NAntC demonstrated low LDH release, and its cytotoxicity remained unaffected by the autophagy inhibitor 3-MA. In an in vivo zebrafish model, 3-NAntC exhibited excellent tolerability, showing no lethal effects and a low rate of malformations at high doses of up to 75 mg/mL. Overall, 3-NAntC emerges as a novel synthetic peptide with promising antitumor effects in vitro against TNBC cells and low toxicity in vivo.
Collapse
|
3533
|
Wang Y, Kong Y, Yang Q, Zhong C, Zhou D. Identification of fibronectin type III domain containing 3B as a potential prognostic and therapeutic target for pancreatic cancer: a preliminary analysis. Eur J Med Res 2024; 29:221. [PMID: 38581008 PMCID: PMC10996089 DOI: 10.1186/s40001-024-01823-6] [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: 01/09/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Fibronectin type III domain containing 3B (FNDC3B), a member of the fibronectin type III domain-containing protein family, has been indicated in various malignancies. However, the precise role of FNDC3B in the progression of pancreatic cancer (PC) still remains to be elucidated. METHODS In this study, we integrated data from the National Center for Biotechnology Information, the Cancer Genome Atlas, Genotype-Tissue Expression database, and Gene Expression Omnibus datasets to analyze FNDC3B expression and its association with various clinicopathological parameters. Subsequently, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, along with Gene Set Enrichment Analysis (GSEA), single sample Gene Set Enrichment Analysis (ssGSEA) and estimate analysis were recruited to delve into the biological function and immune infiltration based on FNDC3B expression. Additionally, the prognostic estimation was conducted using Cox analysis and Kaplan-Meier analysis. Subsequently, a nomogram was constructed according to the result of Cox analysis to enhance the prognostic ability of FNDC3B. Finally, the preliminary biological function of FNDC3B in PC cells was explored. RESULTS The study demonstrated a significantly higher expression of FNDC3B in tumor tissues compared to normal pancreatic tissues, and this expression was significantly associated with various clinicopathological parameters. GSEA revealed the involvement of FNDC3B in biological processes and signaling pathways related to integrin signaling pathway and cell adhesion. Additionally, ssGSEA analysis indicated a positive correlation between FNDC3B expression and infiltration of Th2 cells and neutrophils, while showing a negative correlation with plasmacytoid dendritic cells and Th17 cells infiltration. Kaplan-Meier analysis further supported that high FNDC3B expression in PC patients was linked to shorter overall survival, disease-specific survival, and progression-free interval. However, although univariate analysis demonstrated a significant correlation between FNDC3B expression and prognosis in PC patients, this association did not hold true in multivariate analysis. Finally, our findings highlight the crucial role of FNDC3B expression in regulating proliferation, migration, and invasion abilities of PC cells. CONCLUSION Despite limitations, the findings of this study underscored the potential of FNDC3B as a prognostic biomarker and its pivotal role in driving the progression of PC, particularly in orchestrating immune responses.
Collapse
Affiliation(s)
- Yizhi Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yang Kong
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Qifan Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Cheng Zhong
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Dongkai Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
3534
|
Scott D, Singer DS. Introducing Five New Cancer Grand Challenges Teams. Cancer Discov 2024; 14:559-562. [PMID: 38446429 DOI: 10.1158/2159-8290.cd-24-0153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
SUMMARY Cancer Grand Challenges is an international funding initiative that aims to unite the world's best scientists to tackle some of cancer's toughest problems by funding team science on a global scale. Here, we discuss the five newly funded teams and the challenges they will address over the coming years.
Collapse
Affiliation(s)
- David Scott
- Cancer Grand Challenges, Cancer Research UK, London, United Kingdom
| | | |
Collapse
|
3535
|
Petrella F, Danuzzo F, Sibilia MC, Vaquer S, Longarini R, Guidi A, Raveglia F, Libretti L, Pirondini E, Cara A, Cassina EM, Tuoro A, Cortinovis D. Colorectal Cancer Pulmonary Metastasectomy: When, Why and How. Cancers (Basel) 2024; 16:1408. [PMID: 38611086 PMCID: PMC11010871 DOI: 10.3390/cancers16071408] [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: 03/14/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Colorectal cancer is the third-most-diagnosed cancer in males and in females, representing 8% of estimated new cases, and the third cause of cancer-related death in both sexes, accounting for 9% of cancer deaths in men and 8% in women. About 20% of patients diagnosed with CRC present metastatic disease. Although lung metachronous or synchronous metastatic spread without other involved sites has been reported in only a small proportion of patients, considering that this tumor is frequently diagnosed, the clinical approach to CRC pulmonary metastases represents a major issue for thoracic surgeons and CRC oncologists. Among patients diagnosed with pulmonary metastases from CRC, about 9-12% are eligible for local treatments with radical intent, including surgical resection, SBRT (stereotactic body radiation therapy) and ablation therapy. Due to the lack of randomized controlled trials among different local strategies, there is no definitive evidence about the optimal approach, although surgical resection is considered the most effective therapeutic option in this clinical scenario. Oncological achievement of primary radical resection, the biology of primary tumor and metastatic sites, disease free interval and or progression free survival are independent prognostic factors which make it possible to define a cohort of patients which might significantly benefit from pulmonary metastasectomy.
Collapse
Affiliation(s)
- Francesco Petrella
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (F.D.); (M.C.S.); (S.V.); (L.L.); (E.P.); (A.C.); (E.M.C.); (A.T.)
| | - Federica Danuzzo
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (F.D.); (M.C.S.); (S.V.); (L.L.); (E.P.); (A.C.); (E.M.C.); (A.T.)
| | - Maria Chiara Sibilia
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (F.D.); (M.C.S.); (S.V.); (L.L.); (E.P.); (A.C.); (E.M.C.); (A.T.)
| | - Sara Vaquer
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (F.D.); (M.C.S.); (S.V.); (L.L.); (E.P.); (A.C.); (E.M.C.); (A.T.)
| | - Raffaella Longarini
- Division of Medical Oncology, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (R.L.); or (D.C.)
| | - Alessandro Guidi
- Division of Medical Oncology, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (R.L.); or (D.C.)
| | - Federico Raveglia
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (F.D.); (M.C.S.); (S.V.); (L.L.); (E.P.); (A.C.); (E.M.C.); (A.T.)
| | - Lidia Libretti
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (F.D.); (M.C.S.); (S.V.); (L.L.); (E.P.); (A.C.); (E.M.C.); (A.T.)
| | - Emanuele Pirondini
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (F.D.); (M.C.S.); (S.V.); (L.L.); (E.P.); (A.C.); (E.M.C.); (A.T.)
| | - Andrea Cara
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (F.D.); (M.C.S.); (S.V.); (L.L.); (E.P.); (A.C.); (E.M.C.); (A.T.)
| | - Enrico Mario Cassina
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (F.D.); (M.C.S.); (S.V.); (L.L.); (E.P.); (A.C.); (E.M.C.); (A.T.)
| | - Antonio Tuoro
- Division of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (F.D.); (M.C.S.); (S.V.); (L.L.); (E.P.); (A.C.); (E.M.C.); (A.T.)
| | - Diego Cortinovis
- Division of Medical Oncology, Fondazione IRCCS San Gerardo dei Tintori, Via GB Pergolesi 33, 20900 Monza, Italy; (R.L.); or (D.C.)
- School of Medicine and Surgery, University of Milano Bicocca, 20126 Milan, Italy
| |
Collapse
|
3536
|
Jones AN, Scheurlen KM, Macleod A, Simon HL, Galandiuk S. Obesity and Inflammatory Factors in the Progression of Early-Onset Colorectal Cancer. Cancers (Basel) 2024; 16:1403. [PMID: 38611081 PMCID: PMC11010915 DOI: 10.3390/cancers16071403] [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: 02/23/2024] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
Metabolic dysfunction associated with obesity leads to a chronic pro-inflammatory state with systemic effects, including the alteration of macrophage metabolism. Tumor-associated macrophages have been linked to the formation of cancer through the production of metabolites such as itaconate. Itaconate downregulates peroxisome proliferator-activated receptor gamma as a tumor-suppressing factor and upregulates anti-inflammatory cytokines in M2-like macrophages. Similarly, leptin and adiponectin also influence macrophage cytokine expression and contribute to the progression of colorectal cancer via changes in gene expression within the PI3K/AKT pathway. This pathway influences cell proliferation, differentiation, and tumorigenesis. This work provides a review of obesity-related hormones and inflammatory mechanisms leading to the development and progression of early-onset colorectal cancer (EOCRC). A literature search was performed using the PubMed and Cochrane databases to identify studies related to obesity and EOCRC, with keywords including 'EOCRC', 'obesity', 'obesity-related hormones', 'itaconate', 'adiponectin', 'leptin', 'M2a macrophage', and 'microbiome'. With this concept of pro-inflammatory markers contributing to EOCRC, increased use of chemo-preventative agents such as aspirin may have a protective effect. Elucidating this association between obesity-related, hormone/cytokine-driven inflammatory effects with EOCRC may help lead to new therapeutic targets in preventing and treating EOCRC.
Collapse
Affiliation(s)
- Alexandra N. Jones
- Price Institute of Surgical Research, University of Louisville, Louisville, KY 40202, USA; (A.N.J.); (A.M.); (H.L.S.)
| | - Katharina M. Scheurlen
- Price Institute of Surgical Research, University of Louisville, Louisville, KY 40202, USA; (A.N.J.); (A.M.); (H.L.S.)
| | - Anne Macleod
- Price Institute of Surgical Research, University of Louisville, Louisville, KY 40202, USA; (A.N.J.); (A.M.); (H.L.S.)
| | - Hillary L. Simon
- Price Institute of Surgical Research, University of Louisville, Louisville, KY 40202, USA; (A.N.J.); (A.M.); (H.L.S.)
- Division of Colon and Rectal Surgery, Hiram C. Polk Jr. MD Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| | - Susan Galandiuk
- Price Institute of Surgical Research, University of Louisville, Louisville, KY 40202, USA; (A.N.J.); (A.M.); (H.L.S.)
- Division of Colon and Rectal Surgery, Hiram C. Polk Jr. MD Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
3537
|
Bhandari K, Ding WQ. Protein Arginine Methyltransferases in Pancreatic Ductal Adenocarcinoma: New Molecular Targets for Therapy. Int J Mol Sci 2024; 25:3958. [PMID: 38612768 PMCID: PMC11011826 DOI: 10.3390/ijms25073958] [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: 02/29/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignant disease with a low 5-year overall survival rate. It is the third-leading cause of cancer-related deaths in the United States. The lack of robust therapeutics, absence of effective biomarkers for early detection, and aggressive nature of the tumor contribute to the high mortality rate of PDAC. Notably, the outcomes of recent immunotherapy and targeted therapy against PDAC remain unsatisfactory, indicating the need for novel therapeutic strategies. One of the newly described molecular features of PDAC is the altered expression of protein arginine methyltransferases (PRMTs). PRMTs are a group of enzymes known to methylate arginine residues in both histone and non-histone proteins, thereby mediating cellular homeostasis in biological systems. Some of the PRMT enzymes are known to be overexpressed in PDAC that promotes tumor progression and chemo-resistance via regulating gene transcription, cellular metabolic processes, RNA metabolism, and epithelial mesenchymal transition (EMT). Small-molecule inhibitors of PRMTs are currently under clinical trials and can potentially become a new generation of anti-cancer drugs. This review aims to provide an overview of the current understanding of PRMTs in PDAC, focusing on their pathological roles and their potential as new therapeutic targets.
Collapse
Affiliation(s)
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Sciences Center, BMSB401A, 940 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA;
| |
Collapse
|
3538
|
Sadeghi MH, Sina S, Meigooni AS. Assessing Heterogeneity Effects on Points A, B, and Organs at Risk Doses in High-dose-Rate Brachytherapy for Cervical Cancer - A Comparison of 192Ir and 60Co Sources Using Monte Carlo N-Particle 5. J Med Phys 2024; 49:294-303. [PMID: 39131438 PMCID: PMC11309145 DOI: 10.4103/jmp.jmp_162_23] [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: 11/19/2023] [Revised: 03/24/2024] [Accepted: 04/14/2024] [Indexed: 08/13/2024] Open
Abstract
Purpose The present article deals with investigating the effects of tissue heterogeneity consideration on the dose distribution of 192Ir and 60Co sources in high-dose-rate brachytherapy (HDR-BT). Materials and Methods A Monte Carlo N-Particle 5 (MCNP5) code was developed for the simulation of the dose distribution in homogeneous and heterogeneous phantoms for cervical cancer patients. The phantoms represented water-equivalent and human body-equivalent tissues. Treatment data for a patient undergoing HDR-BT with a 192Ir source were used as a reference for validation, and for 60Co, AAPM Task Group 43 methodology was also applied. The dose values were calculated for both source types in the phantoms. Results The results showed a good agreement between the calculated dose in the homogeneous phantom and the real patient's treatment data, with a relative difference of less than 5% for both sources. However, when comparing the absorbed doses at critical points such as Point A right, Point A left, Point B right, Point B left, bladder International Commission on Radiation Units and Measurement (ICRU) point, and recto-vaginal ICRU point, the study revealed significant percentage differences (approximately 5.85% to 12.02%) between the homogeneous and heterogeneous setups for both 192Ir and 60Co sources. The analysis of dose-volume histograms (DVH) indicated that organs at risk, notably the rectum and bladder, still received doses within recommended limits. Conclusions The study concludes that 60Co and 192Ir sources can be effectively used in HDR-BT, provided that careful consideration is given to tissue heterogeneity effects during treatment planning to ensure optimal therapeutic outcomes.
Collapse
Affiliation(s)
- Mohammad Hossein Sadeghi
- Department of Nuclear Engineering, School of Mechanical Engineering, Shiraz University, Shiraz, Iran
| | - Sedigheh Sina
- Department of Nuclear Engineering, School of Mechanical Engineering, Shiraz University, Shiraz, Iran
- Radiation Research Center, School of Mechanical Engineering, Shiraz University, Shiraz, Iran
| | | |
Collapse
|
3539
|
Nicolae CL, Pîrvulescu DC, Antohi AM, Niculescu AG, Grumezescu AM, Croitoru GA. Silica nanoparticles in medicine: overcoming pathologies through advanced drug delivery, diagnostics, and therapeutic strategies. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2024; 65:173-184. [PMID: 39020531 PMCID: PMC11384868 DOI: 10.47162/rjme.65.2.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
Over the last decades, silica nanoparticles (SiNPs) have been studied for their applications in biomedicine as an alternative used for conventional diagnostics and treatments. Since their properties can be modified and adjusted for the desired use, they have many different potential applications in medicine: they can be used in diagnosis because of their ability to be loaded with dyes and their increased selectivity and sensitivity, which can improve the quality of the diagnostic process. SiNPs can be functionalized by targeting ligands or molecules to detect certain cellular processes or biomarkers with better precision. Targeted delivery is another fundamental use of SiNPs. They could be used as drug delivery systems (DDS) since their structure allows the loading of therapeutic agents or other compounds, and studies have demonstrated their biocompatibility. When SiNPs are used as DDS, the drug's toxicity and the off-target effects are reduced significantly, and they can be used to treat conditions like cancer and neurological diseases and even aid in regenerative processes, such as wound healing or bone repair. However, safety concerns must be considered before SiNPs can be used extensively in clinical practice because NPs can cause toxicity in certain conditions and accumulate at undesired locations. Therefore, an overview of the potential applications that SiNPs could have in medicine, as well as their safety concerns, will be covered in this review paper.
Collapse
Affiliation(s)
- Carmen Larisa Nicolae
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica, Bucharest, Romania;
| | | | | | | | | | | |
Collapse
|
3540
|
Kopelman ZA, Tian C, Tumas J, Phippen NT, Tarney CM, Hope ER, Winkler SS, Jokajtys S, Kucera CW, Chan JK, Richardson MT, Kapp DS, Hamilton CA, Leath CA, Jones NL, Rocconi RP, Farley JH, Secord AA, Cosgrove CM, Powell MA, Klopp A, Walker JL, Fleming GF, Bateman NW, Conrads TP, Maxwell GL, Darcy KM. Disease progression, survival, and molecular disparities in Black and White patients with endometrioid endometrial carcinoma in real-world registries and GOG/NRG oncology randomized phase III clinical trials. Gynecol Oncol 2024; 183:103-114. [PMID: 38593674 PMCID: PMC11894812 DOI: 10.1016/j.ygyno.2024.03.026] [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: 01/17/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024]
Abstract
OBJECTIVE Investigate racial disparities in outcomes and molecular features in Black and White patients with endometrioid endometrial carcinoma (EEC). METHODS Black and White patients diagnosed with EEC who underwent hysterectomy ± adjuvant treatment in SEER, National Cancer Database (NCDB), the Genomics Evidence Neoplasia Information Exchange (GENIE) project (v.13.0), and eight NCI-sponsored randomized phase III clinical trials (RCTs) were studied. Hazard ratio (HR) and 95% confidence interval (CI) were estimated for cancer-related death (CRD), non-cancer death (NCD), and all-cause death. RESULTS Black (n = 4397) vs. White (n = 47,959) patients in SEER had a HR (95% CI) of 2.04 (1.87-2.23) for CRD and 1.22 (1.09-1.36) for NCD. In NCDB, the HR (95% CI) for death in Black (n = 13,468) vs. White (n = 155,706) patients was 1.52 (1.46-1.58) dropping to 1.29 (1.23-1.36) after propensity-score matching for age, comorbidity, income, insurance, grade, stage, LVSI, and treatment. In GENIE, Black (n = 109) vs. White (n = 1780) patients had fewer PTEN, PIK3R1, FBXW7, NF1, mTOR, CCND1, and PI3K-pathway-related gene mutations. In contrast, TP53 and DNA-repair-related gene mutation frequency as well as tumor mutational burden-high status were similar in Black and White patients. In RCTs, Black (n = 187) vs. White (n = 2877) patients were more likely to have advanced or recurrent disease, higher grade, worse performance status and progressive disease. Risk of death in Black vs. White patients in RCTs was 2.19 (1.77-2.71) persisting to 1.32 (1.09-1.61) after matching for grade, stage, and treatment arm while balancing age and performance status. CONCLUSIONS Differences exist in clinical presentation, outcomes, and molecular features in Black vs. White patients with EEC in real-world registries and RCTs. Targeted-drug development, strategies to modify social determinants, and diverse inclusion in RCTs are approaches to reduce disparities.
Collapse
Affiliation(s)
- Zachary A Kopelman
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Chunqiao Tian
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Jordyn Tumas
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Neil T Phippen
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Christopher M Tarney
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Erica R Hope
- Division of Gynecologic Oncology, Department of Gynecologic Surgery and Obstetrics, Brooke Army Medical Center, San Antonio, TX, USA
| | - Stuart S Winkler
- Division of Gynecologic Oncology, Department of Gynecologic Surgery and Obstetrics, Brooke Army Medical Center, San Antonio, TX, USA
| | - Suzanne Jokajtys
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Calen W Kucera
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - John K Chan
- Palo Alto Medical Foundation, California Pacific Medical Center, Sutter Health, San Francisco, CA, USA
| | - Michael T Richardson
- Department of Obstetrics and Gynecology, University of California, Los Angeles School of Medicine, Los Angeles, CA, USA
| | - Daniel S Kapp
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chad A Hamilton
- Gynecologic Oncology Section, Women's Services and The Ochsner Cancer Institute, Ochsner Health, New Orleans, LA, USA
| | - Charles A Leath
- Division of Gynecologic Oncology, University of Alabama at Birmingham, O'Neal Comprehensive Cancer Center, Birmingham, AL, USA
| | - Nathaniel L Jones
- Division of Gynecologic Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Rodney P Rocconi
- Division of Gynecologic Oncology, Cancer Center & Research Institute, the University of Mississippi Medical Center, Jackson, MS, USA
| | - John H Farley
- Division of Gynecologic Oncology, Center for Women's Health, Cancer Institute, Dignity Health St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | | | - Casey M Cosgrove
- Division of Gynecologic Oncology, The Ohio State University, Columbus, OH, USA
| | - Matthew A Powell
- Division of Gynecologic Oncology, Siteman Cancer Center, Washington University, St Louis, MO, USA
| | - Ann Klopp
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joan L Walker
- Gynecologic Oncology Division, Stephenson Cancer Center, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Gini F Fleming
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Nicholas W Bateman
- Division of Gynecologic Oncology, Center for Women's Health, Cancer Institute, Dignity Health St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA; The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Thomas P Conrads
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; Women's Health Integrated Research Center, Inova Women's Service Line, Inova Health System, Falls Church, VA, USA
| | - G Larry Maxwell
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; Women's Health Integrated Research Center, Inova Women's Service Line, Inova Health System, Falls Church, VA, USA.
| | - Kathleen M Darcy
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA.
| |
Collapse
|
3541
|
Soltan A, Washington P. Challenges in Reducing Bias Using Post-Processing Fairness for Breast Cancer Stage Classification with Deep Learning. ALGORITHMS 2024; 17:141. [PMID: 38962581 PMCID: PMC11221567 DOI: 10.3390/a17040141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Breast cancer is the most common cancer affecting women globally. Despite the significant impact of deep learning models on breast cancer diagnosis and treatment, achieving fairness or equitable outcomes across diverse populations remains a challenge when some demographic groups are underrepresented in the training data. We quantified the bias of models trained to predict breast cancer stage from a dataset consisting of 1000 biopsies from 842 patients provided by AIM-Ahead (Artificial Intelligence/Machine Learning Consortium to Advance Health Equity and Researcher Diversity). Notably, the majority of data (over 70%) were from White patients. We found that prior to post-processing adjustments, all deep learning models we trained consistently performed better for White patients than for non-White patients. After model calibration, we observed mixed results, with only some models demonstrating improved performance. This work provides a case study of bias in breast cancer medical imaging models and highlights the challenges in using post-processing to attempt to achieve fairness.
Collapse
Affiliation(s)
- Armin Soltan
- Hawaii Health Digital Lab, Information and Computer Science, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Peter Washington
- Hawaii Health Digital Lab, Information and Computer Science, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| |
Collapse
|
3542
|
Xu J, Zhang Z, Hu H, Yang Y, Xiao C, Xi L, Lu J, Tian S, Zhao H. Synergistic antitumor effects of Peiminine and Doxorubicin on breast cancer through enhancing DNA damage via ZEB1. Biomed Pharmacother 2024; 173:116353. [PMID: 38432128 DOI: 10.1016/j.biopha.2024.116353] [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: 12/20/2023] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
Peiminine, the primary biologically active compound from Fritillaria thunbergii Miq., has demonstrated significant pharmacological activities. Doxorubicin is one of the most potent chemotherapeutic agents for breast cancer (BC). This study was designed to investigate the efficacy and underlying mechanisms of Peiminine combined with Doxorubicin in treating BC. Our results demonstrated that the combination of Peiminine and 1 mg/kg Doxorubicin exhibited more significant suppression of tumor growth compared with the monotherapy in MDA-MB-231 xenograft nude mice model, which is comparable to the effect of 3 mg/kg Doxorubicin in vivo. Notably, the 3 mg/kg Doxorubicin monotherapy resulted in organ toxicity, specifically in the liver and heart, whereas no toxicity was observed in the combination group. In vitro, this combined treatment exhibited a synergistic reduction on the viability of BC cells. Peiminine enhanced the cell cycle arrest and DNA damage induced by Doxorubicin. Furthermore, the combination treatment effectively blocked DNA repair by inhibiting the MAPKs signaling pathways. And ZEB1 knockdown attenuated the combined effect of Peiminine and Doxorubicin on cell viability and DNA damage. In conclusion, our study found that the combination of Peiminine and Doxorubicin showed synergistic inhibitory effects on BC both in vivo and in vitro through enhancing Doxorubicin-induced DNA damage. These findings support that their combination is a novel and promising therapeutic strategy for treating BC.
Collapse
Affiliation(s)
- Jiajin Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China
| | - Zeyi Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China
| | - Hongtao Hu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China
| | - Yaqin Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China
| | - Chenghong Xiao
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, China
| | - Luyi Xi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China
| | - Jiahui Lu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China
| | - Shasha Tian
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China.
| | - Huajun Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Binwen Rd., Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
3543
|
Wang Q, Zhang C, Qi C, Qiang Y, Zhang Z, Xu F, Shen Y. Esophageal surgical Apgar score (eSAS): A predictor for postoperative morbidity in patients undergoing neoadjuvant therapy and esophagectomy. Thorac Cancer 2024; 15:755-763. [PMID: 38390683 PMCID: PMC10995716 DOI: 10.1111/1759-7714.15246] [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: 11/24/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND The surgical Apgar score (SAS) quantifying three intraoperative indexes has been confirmed to be significantly associated with postoperative morbidity and prognosis in many surgical specialties. However, there are great limitations in its application for esophageal cancer (EC). This study aimed to assess the predictive capability of esophagectomy SAS (eSAS) in determining postoperative morbidity and overall survival (OS) in EC patients who had undergone neoadjuvant therapy. METHODS A retrospective evaluation was conducted on a cohort of 221 patients in which surgery- and tumor-related data were extracted and analyzed. Major morbidity was defined as complications meeting the criteria of Clavien-Dindo classification III or higher during hospitalization. Univariate and multivariate analyses were performed to identify potential risk factors for major morbidity. Kaplan-Meier analysis was utilized to calculate the OS and relapse-free survival (RFS). RESULTS The results exhibited that eSAS demonstrated potential predictive value for postoperative morbidity with an optimal cutoff value of 6. The eSAS and diabetes mellitus were two independent risk factors for the major morbidity; however, no correlation between the eSAS and the OS or RFS was detected. CONCLUSION The eSAS could be used as a predictor of major morbidity, while it was not correlated with OS and RFS.
Collapse
Affiliation(s)
- Qin Wang
- Department of Cardiothoracic Surgery, Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Chi Zhang
- Department of Cardiothoracic Surgery, Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Chen Qi
- Department of Cardiothoracic Surgery, Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Yong Qiang
- Department of Cardiothoracic Surgery, Jinling Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Zheng Zhang
- Department of Cardiothoracic Surgery, Jinling Hospital, School of Clinical MedicineNanjing Medical UniversityNanjingChina
| | - Fei Xu
- Department of Cardiothoracic Surgery, Jinling Hospital, School of Clinical MedicineNanjing Medical UniversityNanjingChina
| | - Yi Shen
- Department of Cardiothoracic Surgery, Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| |
Collapse
|
3544
|
Lloyd EG, Henríquez JA, Biffi G. Modelling the micro- and macro- environment of pancreatic cancer: from patients to pre-clinical models and back. Dis Model Mech 2024; 17:dmm050624. [PMID: 38639944 PMCID: PMC11051978 DOI: 10.1242/dmm.050624] [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: 04/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with very low survival rates. Over the past 50 years, improvements in PDAC survival have significantly lagged behind the progress made in other cancers. PDAC's dismal prognosis is due to typical late-stage diagnosis combined with lack of effective treatments and complex mechanisms of disease. We propose that improvements in survival are partly hindered by the current focus on largely modelling and targeting PDAC as one disease, despite it being heterogeneous. Implementing new disease-representative pre-clinical mouse models that capture this complexity could enable the development of transformative therapies. Specifically, these models should recapitulate human PDAC late-stage biology, heterogeneous genetics, extensive non-malignant stroma, and associated risk factors and comorbidities. In this Perspective, we focus on how pre-clinical mouse models could be improved to exemplify key features of PDAC micro- and macro- environments, which would drive clinically relevant patient stratification, tailored treatments and improved survival.
Collapse
Affiliation(s)
- Eloise G. Lloyd
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| | - Joaquín Araos Henríquez
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| | - Giulia Biffi
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
3545
|
Massey A, Stewart J, Smith C, Parvini C, McCormick M, Do K, Cartagena-Rivera AX. Mechanical properties of human tumour tissues and their implications for cancer development. NATURE REVIEWS. PHYSICS 2024; 6:269-282. [PMID: 38706694 PMCID: PMC11066734 DOI: 10.1038/s42254-024-00707-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 05/07/2024]
Abstract
The mechanical properties of cells and tissues help determine their architecture, composition and function. Alterations to these properties are associated with many diseases, including cancer. Tensional, compressive, adhesive, elastic and viscous properties of individual cells and multicellular tissues are mostly regulated by reorganization of the actomyosin and microtubule cytoskeletons and extracellular glycocalyx, which in turn drive many pathophysiological processes, including cancer progression. This Review provides an in-depth collection of quantitative data on diverse mechanical properties of living human cancer cells and tissues. Additionally, the implications of mechanical property changes for cancer development are discussed. An increased knowledge of the mechanical properties of the tumour microenvironment, as collected using biomechanical approaches capable of multi-timescale and multiparametric analyses, will provide a better understanding of the complex mechanical determinants of cancer organization and progression. This information can lead to a further understanding of resistance mechanisms to chemotherapies and immunotherapies and the metastatic cascade.
Collapse
Affiliation(s)
- Andrew Massey
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Jamie Stewart
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- These authors contributed equally: Jamie Stewart, Chynna Smith
| | - Chynna Smith
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- These authors contributed equally: Jamie Stewart, Chynna Smith
| | - Cameron Parvini
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Moira McCormick
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Kun Do
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Alexander X. Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
3546
|
Wang Y, Kong Y, Yang Q, Zhong C, Zhou D, Wang W. Survival benefit of adjuvant chemotherapy in patients with resected gallbladder adenocarcinoma: An updated retrospective cohort analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108047. [PMID: 38401352 DOI: 10.1016/j.ejso.2024.108047] [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: 12/29/2023] [Revised: 02/11/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND The rarity yet high malignancy of gallbladder adenocarcinoma (GBA) endows it with a distinctive nature. Radical resection remains the foremost therapeutic approach for GBA, while the impact of early recurrence and metastasis on patient prognosis necessitates the utilization of adjuvant chemotherapy (AC). Despite numerous previous studies on this topic, a consensus regarding the authentic efficacy of AC has yet to be reached. METHODS We conducted an updated retrospective cohort analysis utilizing data from the Surveillance, Epidemiology, and End Results (SEER) database spanning from 2010 to 2020 to explore the association between AC and survival outcomes in patients with resected GBA. RESULTS Our study included 2782 patients from the SEER database, with further evaluation of 843 patients in each cohort following meticulous execution of a 1:1 propensity score matching. Remarkably, the AC cohort exhibited a significant survival advantage when juxtaposed against the non-AC cohort. Multivariable Cox regression analysis identified age at diagnosis, year at diagnosis, grade, AJCC T stage, AJCC N stage as well as AC as independent prognostic factors. Furthermore, our findings unveiled that poor/undifferentiated tumor histology, pathological T2 or higher category and pathological N1 category were significantly associated with improved survival when treated with AC while simultaneously observing improved survival across all age categories. CONCLUSION These results provide additional evidence supporting the survival benefits of AC and offer guidance for personalized therapy in patients with resected GBA.
Collapse
Affiliation(s)
- Yizhi Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, 310009, China; Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, 310009, China; Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Yang Kong
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, 310009, China; Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, 310009, China; Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Qifan Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, 310009, China; Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, 310009, China; Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Cheng Zhong
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, 310009, China; Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, 310009, China; Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Dongkai Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, 310009, China; Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, 310009, China; Cancer Center, Zhejiang University, Hangzhou, 310009, China.
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, 310009, China; Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, 310009, China; Cancer Center, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
3547
|
Ye J, Liu F, Zhang L, Wu C, Jiang A, Xie T, Jiang H, Li Z, Luo P, Jiao J, Xiao J. MOCS, a novel classifier system integrated multimoics analysis refining molecular subtypes and prognosis for skin melanoma. J Biomol Struct Dyn 2024:1-17. [PMID: 38555737 DOI: 10.1080/07391102.2024.2329305] [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: 08/29/2023] [Accepted: 02/08/2024] [Indexed: 04/02/2024]
Abstract
PURPOSE The present investigation focuses on Skin Cutaneous Melanoma (SKCM), a melanocytic carcinoma characterized by marked aggression, significant heterogeneity, and a complex etiological background, factors which collectively contribute to the challenge in prognostic determinations. We defined a novel classifier system specifically tailored for SKCM based on multiomics. METHODS We collected 423 SKCM samples with multi omics datasets to perform a consensus cluster analysis using 10 machine learning algorithms and verified in 2 independent cohorts. Clinical features, biological characteristics, immune infiltration pattern, therapeutic response and mutation landscape were compared between subtypes. RESULTS Based on consensus clustering algorithms, we identified two Multi-Omics-Based-Cancer-Subtypes (MOCS) in SKCM in TCGA project and validated in GSE19234 and GSE65904 cohorts. MOCS2 emerged as a subtype with poor prognosis, characterized by a complex immune microenvironment, dysfunctional anti-tumor immune state, high cancer stemness index, and genomic instability. MOCS2 exhibited resistance to chemotherapy agents like erlotinib and sunitinib while sensitive to rapamycin, NSC87877, MG132, and FH355. Additionally, ELSPBP1 was identified as the target involving in glycolysis and M2 macrophage infiltration in SKCM. CONCLUSIONS MOCS classification could stably predict prognosis of SKCM; patients with a high cancer stemness index combined with genomic instability may be predisposed to an immune exhaustion state.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Juelan Ye
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University (Second Military Medical University), Shanghai, China
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Fuchun Liu
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University (Second Military Medical University), Shanghai, China
| | - Luoshen Zhang
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University (Second Military Medical University), Shanghai, China
| | - Chunbiao Wu
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University (Second Military Medical University), Shanghai, China
- School of Health Science and Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Tianying Xie
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University (Second Military Medical University), Shanghai, China
- School of Health Science and Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Hao Jiang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University (Second Military Medical University), Shanghai, China
- School of Health Science and Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Zhenxi Li
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University (Second Military Medical University), Shanghai, China
- School of Health Science and Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Jiao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jianru Xiao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University (Second Military Medical University), Shanghai, China
- School of Health Science and Technology, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
3548
|
Padasdao B, Konh B. A mechanics-based model for a tendon-driven active needle navigating inside a multiple-layer tissue. J Robot Surg 2024; 18:146. [PMID: 38554177 PMCID: PMC11034936 DOI: 10.1007/s11701-024-01900-2] [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: 02/08/2024] [Accepted: 03/02/2024] [Indexed: 04/01/2024]
Abstract
Percutaneous minimally invasive procedures such brachytherapy and biopsy require a flexible active needle for precise movement inside tissue and accurate placement at target positions for higher success rates for diagnosis and treatment, respectively. In a previous work, we presented a tendon-driven active needle to navigate inside tissue. This work presents a new model to predict the deflection of the tendon-driven needle while steering in a multiple-layer soft tissue. A multi-layer phantom tissue with different localized stiffness was developed for needle insertion tests followed by indentation tests to identify its mechanical properties. Using a robot that inserts and actively bends the tendon-driven needle inside the soft tissue while simultaneously tracking the needle through ultrasound imaging, various experiments were conducted for model validation. The proposed model was verified by comparing the simulation results to the empirical data. The results demonstrated the accuracy of the model in predicting the tendon-driven needle deflection in multiple-layer (different stiffness) soft tissue.
Collapse
Affiliation(s)
| | - Bardia Konh
- University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
3549
|
Ghazal LV, Johnston H, Dodd E, Ramachandra Y, Giallourakis N, Fulginiti K, Kamen C. A Needs Assessment Approach for Adolescent and Young Adult Sexual and Gender Diverse Cancer Survivors. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:424. [PMID: 38673335 PMCID: PMC11050404 DOI: 10.3390/ijerph21040424] [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: 02/25/2024] [Revised: 03/22/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024]
Abstract
Sexual and gender diverse (SGD) adolescent and young adult (AYA) cancer survivors are an increasing and vulnerable group with unique needs that often remain unmet in the healthcare system. This paper describes the conceptualization and development of a community-based organization dedicated to serving SGD AYAs, in addition to reporting on the results of a community-led needs assessment. A total of 56 SGD AYA community members completed the online survey. Most participants were between the ages of 26 to 33, identified as white, cisgender, bisexual women, and had hematologic malignancies. Identified unmet needs of SGD AYAs included the following: sexual health and family planning; gender affirmation; financial stability; and emotional support. Areas within the community organization were identified as gaps, areas of expansion, and assets. Results highlight the role of community and academic partnerships in improving cancer care delivery for SGD AYA cancer survivors.
Collapse
Affiliation(s)
- Lauren V. Ghazal
- School of Nursing, University of Rochester, Rochester, NY 14642, USA
- Wilmot Cancer Institute, Rochester, NY 14642, USA;
| | - Hailey Johnston
- Escape, Lansing, MI 48915, USA; (H.J.); (E.D.); (Y.R.)
- School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | - Charles Kamen
- Wilmot Cancer Institute, Rochester, NY 14642, USA;
- Department of Surgery, Cancer Control, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
3550
|
Fumagalli D, De Vitis LA, Caruso G, Occhiali T, Palmieri E, Guillot BE, Pappalettera G, Langstraat CL, Glaser GE, Reynolds EA, Fruscio R, Landoni F, Mariani A, Grassi T. Low-Volume Metastases in Apparent Early-Stage Endometrial Cancer: Prevalence, Clinical Significance, and Future Perspectives. Cancers (Basel) 2024; 16:1338. [PMID: 38611016 PMCID: PMC11011093 DOI: 10.3390/cancers16071338] [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: 02/27/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Endometrial cancer (EC) is the most diagnosed gynecologic malignancy, and its incidence and mortality are increasing. The prognosis is highly dependent on the disease spread. Surgical staging includes retroperitoneal evaluation to detect potential lymph node metastases. In recent years, systematic lymphadenectomy has been replaced by sentinel lymph node (SLN) biopsy and ultrastaging, allowing for the detection of macrometastases, micrometastases, and isolated tumor cells (ITCs). Micrometastases and ITCs have been grouped as low-volume metastases (LVM). The reported prevalence of LVM in studies enrolling more than one thousand patients with apparent early-stage EC ranges from 1.9% to 10.2%. Different rates of LVM are observed when patients are stratified according to disease characteristics and their risk of recurrence. Patients with EC at low risk for recurrence have low rates of LVM, while intermediate- and high-risk patients have a higher likelihood of being diagnosed with nodal metastases, including LVM. Macro- and micrometastases increase the risk of recurrence and cause upstaging, while the clinical significance of ITCs is still uncertain. A recent meta-analysis found that patients with LVM have a higher relative risk of recurrence [1.34 (95% CI: 1.07-1.67)], regardless of adjuvant treatment. In a retrospective study on patients with low-risk EC and no adjuvant treatment, those with ITCs had worse recurrence-free survival compared to node-negative patients (85.1%; CI 95% 73.8-98.2 versus 90.2%; CI 95% 84.9-95.8). However, a difference was no longer observed after the exclusion of cases with lymphovascular space invasion. There is no consensus on adjuvant treatment in ITC patients at otherwise low risk, and their recurrence rate is low. Multi-institutional, prospective studies are warranted to evaluate the clinical significance of ITCs in low-risk patients. Further stratification of patients, considering histopathological and molecular features of the disease, may clarify the role of LVM and especially ITCs in specific contexts.
Collapse
Affiliation(s)
- Diletta Fumagalli
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN 55905, USA; (D.F.); (L.A.D.V.); (G.C.); (T.O.); (E.P.); (B.E.G.); (G.P.); (C.L.L.); (G.E.G.); (E.A.R.); (A.M.)
- Department of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (R.F.); (F.L.)
| | - Luigi A. De Vitis
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN 55905, USA; (D.F.); (L.A.D.V.); (G.C.); (T.O.); (E.P.); (B.E.G.); (G.P.); (C.L.L.); (G.E.G.); (E.A.R.); (A.M.)
- Department of Gynecology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
| | - Giuseppe Caruso
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN 55905, USA; (D.F.); (L.A.D.V.); (G.C.); (T.O.); (E.P.); (B.E.G.); (G.P.); (C.L.L.); (G.E.G.); (E.A.R.); (A.M.)
- Department of Gynecology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
| | - Tommaso Occhiali
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN 55905, USA; (D.F.); (L.A.D.V.); (G.C.); (T.O.); (E.P.); (B.E.G.); (G.P.); (C.L.L.); (G.E.G.); (E.A.R.); (A.M.)
- Clinic of Obstetrics and Gynecology, Santa Maria della Misericordia University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Emilia Palmieri
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN 55905, USA; (D.F.); (L.A.D.V.); (G.C.); (T.O.); (E.P.); (B.E.G.); (G.P.); (C.L.L.); (G.E.G.); (E.A.R.); (A.M.)
- Gynecologic Oncology Unit, Department of Women, Children and Public Health Sciences, , Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00136 Roma, Italy
| | - Benedetto E. Guillot
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN 55905, USA; (D.F.); (L.A.D.V.); (G.C.); (T.O.); (E.P.); (B.E.G.); (G.P.); (C.L.L.); (G.E.G.); (E.A.R.); (A.M.)
- Department of Gynecology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
| | - Giulia Pappalettera
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN 55905, USA; (D.F.); (L.A.D.V.); (G.C.); (T.O.); (E.P.); (B.E.G.); (G.P.); (C.L.L.); (G.E.G.); (E.A.R.); (A.M.)
- Department of Gynecology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
| | - Carrie L. Langstraat
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN 55905, USA; (D.F.); (L.A.D.V.); (G.C.); (T.O.); (E.P.); (B.E.G.); (G.P.); (C.L.L.); (G.E.G.); (E.A.R.); (A.M.)
| | - Gretchen E. Glaser
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN 55905, USA; (D.F.); (L.A.D.V.); (G.C.); (T.O.); (E.P.); (B.E.G.); (G.P.); (C.L.L.); (G.E.G.); (E.A.R.); (A.M.)
| | - Evelyn A. Reynolds
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN 55905, USA; (D.F.); (L.A.D.V.); (G.C.); (T.O.); (E.P.); (B.E.G.); (G.P.); (C.L.L.); (G.E.G.); (E.A.R.); (A.M.)
| | - Robert Fruscio
- Department of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (R.F.); (F.L.)
- Division of Gynecologic Surgery, IRCCS Fondazione San Gerardo dei Tintori, 20900 Monza, Italy
| | - Fabio Landoni
- Department of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (R.F.); (F.L.)
- Division of Gynecologic Surgery, IRCCS Fondazione San Gerardo dei Tintori, 20900 Monza, Italy
| | - Andrea Mariani
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN 55905, USA; (D.F.); (L.A.D.V.); (G.C.); (T.O.); (E.P.); (B.E.G.); (G.P.); (C.L.L.); (G.E.G.); (E.A.R.); (A.M.)
| | - Tommaso Grassi
- Department of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (R.F.); (F.L.)
| |
Collapse
|