201
|
Hwang HJ, Han SA, Sohn IS. Breast Cancer and Therapy-Related Cardiovascular Toxicity. J Breast Cancer 2024; 27:147-162. [PMID: 38769686 PMCID: PMC11221208 DOI: 10.4048/jbc.2024.0085] [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: 04/02/2024] [Revised: 04/10/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024] Open
Abstract
The global incidence of breast cancer is on the rise, a trend also observed in South Korea. However, thanks to the rapid advancements in anticancer therapies, survival rates are improving. Consequently, post-treatment health and quality of life for breast cancer survivors are emerging as significant concerns, particularly regarding treatment-related cardiotoxicity. In this review, we delve into the cardiovascular complications associated with breast cancer treatment, explore surveillance protocols for early detection and diagnosis of late complications, and discuss protective strategies against cardiotoxicity in breast cancer patients undergoing anticancer therapy, drawing from multiple guidelines.
Collapse
Affiliation(s)
- Hui-Jeong Hwang
- Department of Cardiology, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Sang-Ah Han
- Department of Surgery, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Il Suk Sohn
- Department of Cardiology, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Korea.
| |
Collapse
|
202
|
Marok FZ, Wojtyniak JG, Selzer D, Dallmann R, Swen JJ, Guchelaar HJ, Schwab M, Lehr T. Personalized Chronomodulated 5-Fluorouracil Treatment: A Physiologically-Based Pharmacokinetic Precision Dosing Approach for Optimizing Cancer Therapy. Clin Pharmacol Ther 2024; 115:1282-1292. [PMID: 38264789 DOI: 10.1002/cpt.3181] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/03/2024] [Indexed: 01/25/2024]
Abstract
The discovery of circadian clock genes greatly amplified the study of diurnal variations impacting cancer therapy, transforming it into a rapidly growing field of research. Especially, use of chronomodulated treatment with 5-fluorouracil (5-FU) has gained significance. Studies indicate high interindividual variability (IIV) in diurnal variations in dihydropyrimidine dehydrogenase (DPD) activity - a key enzyme for 5-FU metabolism. However, the influence of individual DPD chronotypes on chronomodulated therapy remains unclear and warrants further investigation. To optimize precision dosing of chronomodulated 5-FU, this study aims to: (i) build physiologically-based pharmacokinetic (PBPK) models for 5-FU, uracil, and their metabolites, (ii) assess the impact of diurnal variation on DPD activity, (iii) estimate individual DPD chronotypes, and (iv) personalize chronomodulated 5-FU infusion rates based on a patient's DPD chronotype. Whole-body PBPK models were developed with PK-Sim(R) and MoBi(R). Sinusoidal functions were used to incorporate variations in enzyme activity and chronomodulated infusion rates as well as to estimate individual DPD chronotypes from DPYD mRNA expression or DPD enzymatic activity. Four whole-body PBPK models for 5-FU, uracil, and their metabolites were established utilizing data from 41 5-FU and 10 publicly available uracil studies. IIV in DPD chronotypes was assessed and personalized chronomodulated administrations were developed to achieve (i) comparable 5-FU peak plasma concentrations, (ii) comparable 5-FU exposure, and (iii) constant 5-FU plasma levels via "noise cancellation" chronomodulated infusion. The developed PBPK models capture the extent of diurnal variations in DPD activity and can help investigate individualized chronomodulated 5-FU therapy through testing alternative personalized dosing strategies.
Collapse
Affiliation(s)
| | - Jan-Georg Wojtyniak
- Clinical Pharmacy, Saarland University, Saarbruecken, Germany
- Dr. Margarete Fischer-Bosch-Institut of Clinical Pharmacology, Stuttgart, Germany
| | - Dominik Selzer
- Clinical Pharmacy, Saarland University, Saarbruecken, Germany
| | - Robert Dallmann
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Jesse J Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, RC Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, RC Leiden, The Netherlands
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institut of Clinical Pharmacology, Stuttgart, Germany
- Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University Tuebingen, Tuebingen, Germany
- Cluster of excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University Tuebingen, Tuebingen, Germany
| | - Thorsten Lehr
- Clinical Pharmacy, Saarland University, Saarbruecken, Germany
| |
Collapse
|
203
|
Zhang X, Zhou L, Zhou C, Shen L. Real-World Effectiveness and Safety of Ramucirumab as a Second-Line Treatment for Patients with Unresectable Advanced or Metastatic Gastric/Gastroesophageal Junction Adenocarcinoma in Japan and South Korea: A Systematic Literature Review. Adv Ther 2024; 41:2112-2132. [PMID: 38619719 PMCID: PMC11133076 DOI: 10.1007/s12325-024-02838-5] [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: 12/14/2023] [Accepted: 03/08/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Gastric cancer has the highest incidence and mortality in Eastern Asia. The efficacy and safety of ramucirumab (RAM) monotherapy or in combination with paclitaxel (PTX) for patients with unresectable advanced or metastatic gastric/gastroesophageal junction adenocarcinoma (G/GEA) have been established in clinical trials. To assess the effectiveness and safety of RAM or RAM-based therapy as a second-line treatment in real-world clinical practice in Eastern Asia and to pave the way for future research, a systematic literature review (SLR) was conducted. METHODS Studies published between January 2014 and December 2021 were identified in PubMed, Embase, Cochrane Library, CNKI, Wanfang, and CBM databases. RESULTS This SLR included 23 studies from Japan and South Korea, of which 22 were retrospective and 11 were full-text articles. Most studies investigated RAM + PTX (range of median overall survival [mOS] 7.4-12.2 months; median progression-free survival [mPFS] 3.35-7.0 months). Data were limited for RAM, RAM + albumin-bound paclitaxel, and RAM + taxane. RAM + PTX was associated with longer survival (mOS 9.3-12.2 months vs. 5.2-9.7 months; mPFS 4.1-5.1 months vs. 3.0-4.1 months) than PTX. Patients with prior anti-programmed cell death 1 (anti-PD-1) exposure experienced longer mPFS (4.8 vs. 3.4 months) from RAM + taxane than those without prior anti-PD-1 exposure. Few patients (3.3-6.3%) discontinued RAM or RAM-based therapy because of adverse events (AEs). Hematological toxicities were most frequently occurring AEs and no new safety signals were identified compared to clinical trials. CONCLUSION RAM + PTX as a second-line treatment is effective and associated with an acceptable toxicity profile in patients with advanced or metastatic G/GEA in real-world settings of Japan and South Korea. More studies are recommended to further evaluate effectiveness and safety of RAM or RAM-based therapy, especially after anti-PD-1 therapy, in a wider Eastern Asian population. TRIAL REGISTRATION INPLASY registration number INPLASY2022120023.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Li Zhou
- Eli Lilly and Company, Shanghai, China
| | - Chan Zhou
- Eli Lilly and Company, Shanghai, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
204
|
Murai S, Nozawa H, Yamada K, Saiki Y, Sasaki K, Murono K, Emoto S, Matsuzaki H, Yokoyama Y, Abe S, Nagai Y, Yoshioka Y, Shinagawa T, Sonoda H, Sugihara K, Ajioka Y, Ishihara S. Local excision versus radical surgery for anal squamous cell carcinoma: a multicenter study in Japan. Int J Clin Oncol 2024; 29:813-821. [PMID: 38526623 PMCID: PMC11130036 DOI: 10.1007/s10147-024-02498-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: 10/22/2023] [Accepted: 02/21/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND The standard treatment for anal squamous cell carcinoma is chemoradiation therapy (CRT), but there is a possibility of over-treatment for early-stage disease. cTisN0 and cT1N0 disease is currently indicated for local excision, but it is unclear whether the indication of local excision can be expanded to cT2N0 disease. METHODS 126 patients with cTis-T2N0 anal cancer treated at 47 centers in Japan between 1991 and 2015 were included. Patients were first classified into the CRT group and surgical therapy group according to the initial therapy, and the latter was further divided into local excision (LE) and radical surgery (RS) groups. We compared prognoses among the groups, and analyzed risk factors for recurrence after local excision. RESULTS The CRT group (n = 87) and surgical therapy group (n = 39) showed no difference in relapse-free survival (p = 0.29) and overall survival (p = 0.94). Relapse-free survival curves in the LE (n = 23) and RS groups (n = 16) overlapped for the initial 3 years, but the curve for the LE group went lower beyond (p = 0.33). By contrast, there was no difference in overall survival between the two groups (p = 0.98). In the LE group, the majority of recurrences distributed in locoregional areas, which could be managed by salvage treatments. Muscular invasion was associated with recurrence after local excision (hazard ratio: 22.91, p = 0.011). CONCLUSION LE may be applied to selected patients with anal cancer of cTis-T2N0 stage. Given the high risk of recurrence in cases with muscular invasion, it may be important to consider close surveillance and additional treatment in such patients.
Collapse
Affiliation(s)
- Shin Murai
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
- Project Committee for Handling Anal Cancers, The Japanese Society for Cancer of the Colon and Rectum, Tokyo, Japan
| | - Hiroaki Nozawa
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan.
- Project Committee for Handling Anal Cancers, The Japanese Society for Cancer of the Colon and Rectum, Tokyo, Japan.
| | - Kazutaka Yamada
- Department of Surgery, Takano Hospital, Kumamoto, Japan
- Project Committee for Handling Anal Cancers, The Japanese Society for Cancer of the Colon and Rectum, Tokyo, Japan
| | - Yasumitsu Saiki
- Department of Surgery, Takano Hospital, Kumamoto, Japan
- Project Committee for Handling Anal Cancers, The Japanese Society for Cancer of the Colon and Rectum, Tokyo, Japan
| | - Kazuhito Sasaki
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Koji Murono
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Shigenobu Emoto
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Hiroyuki Matsuzaki
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Yuichiro Yokoyama
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Shinya Abe
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Yuzo Nagai
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Yuichiro Yoshioka
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Takahide Shinagawa
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Hirofumi Sonoda
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Kenichi Sugihara
- Tokyo Medical and Dental University, Tokyo, Japan
- Project Committee for Handling Anal Cancers, The Japanese Society for Cancer of the Colon and Rectum, Tokyo, Japan
| | - Yoichi Ajioka
- Division of Molecular and Diagnostic Pathology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Project Committee for Handling Anal Cancers, The Japanese Society for Cancer of the Colon and Rectum, Tokyo, Japan
| | - Soichiro Ishihara
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
- Project Committee for Handling Anal Cancers, The Japanese Society for Cancer of the Colon and Rectum, Tokyo, Japan
| |
Collapse
|
205
|
Liu H, Chen J, Huang Y, Zhang Y, Ni Y, Xu N, Zhao F, Tang Y, Liu H, Sun G, Shen P, Liu Z, Huang J, Liao B, Zeng H. Prognostic significance of circulating tumor DNA in urothelial carcinoma: a systematic review and meta-analysis. Int J Surg 2024; 110:3923-3936. [PMID: 38573063 PMCID: PMC11175790 DOI: 10.1097/js9.0000000000001372] [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: 10/27/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) has emerged as a noninvasive technique that provides valuable insights into molecular profiles and tumor disease management. This study aimed to evaluate the prognostic significance of circulating tumor DNA (ctDNA) in urothelial carcinoma (UC) through a systematic review and meta-analysis. METHODS A comprehensive search was conducted in MEDLINE, EMBASE, and the Cochrane Library from the inception to December 2023. Studies investigating the prognostic value of ctDNA in UC were included. Hazard ratios (HRs) of disease-free survival (DFS) and overall survival (OS) were extracted. Overall meta-analysis and subgroup exploration stratified by metastatic status, ctDNA sampling time, treatment type, and detection method was performed using the R software (version 4.2.2). RESULTS A total of 16 studies with 1725 patients were included. Fourteen studies assessed the association between baseline ctDNA status and patient outcomes. Patients with elevated ctDNA levels exhibited significantly worse DFS (HR=6.26; 95% CI: 3.71-10.58, P <0.001) and OS (HR=4.23; 95% CI: 2.72-6.57, P <0.001) regardless of metastatic status, ctDNA sampling time, treatment type, and detection methods. Six studies evaluated the prognostic value of ctDNA dynamics in UC. Patients who showed a decrease or clearance in ctDNA levels during treatment or observation demonstrated more favorable DFS (HR=0.26, 95% CI: 0.17-0.41, P <0.001) and OS (HR=0.21, 95% CI: 0.11-0.38, P <0.001) compared to those who did not. The association remained consistent across the subgroup analysis based on metastatic status and detection methods. In the immune checkpoint inhibitor-treated setting, both lower baseline ctDNA level and ctDNA decrease during the treatment were significantly associated with more favorable oncologic outcomes. Furthermore, specific gene mutations such as FGFR3 identified in ctDNA also demonstrated predictive value in UC patients. CONCLUSION This meta-analysis demonstrates a strong association of ctDNA status and its dynamic change with survival outcomes in UC, suggesting substantial clinical utility of ctDNA testing in prognosis prediction and decision making in this setting.
Collapse
Affiliation(s)
- Haoyang Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Junru Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Yuchen Huang
- Department of Cardiothoracic Surgery, West China Hospital, Sichuan University
| | - Yaowen Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Yuchao Ni
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Nanwei Xu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Fengnian Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Yanfeng Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Haolin Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Guangxi Sun
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Pengfei Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Jin Huang
- Medical Device Regulatory Research and Evaluation Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Banghua Liao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| |
Collapse
|
206
|
Moten AS. Fibrosis following neoadjuvant treatment of PDAC: Less is not always more. Am J Surg 2024; 232:8. [PMID: 38092641 DOI: 10.1016/j.amjsurg.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 05/15/2024]
Affiliation(s)
- Ambria S Moten
- Surgical Oncology, Complex Upper GI/HPB Surgery, Packnett Family Cancer Institute, Parkview Health, 11050 Parkview Circle, Fort Wayne, IN, USA.
| |
Collapse
|
207
|
Kei C, Gartrell R, Arafat Y, Degabriele E, Yeung J, Chan S, Faragher I, Yeung JMC. Colorectal cancer treatment outcomes during the pandemic: Our experience of COVID-19 at a tertiary referral center. Asia Pac J Clin Oncol 2024; 20:395-406. [PMID: 38391122 DOI: 10.1111/ajco.14051] [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: 04/29/2023] [Revised: 11/12/2023] [Accepted: 02/06/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUNDS The coronavirus disease 2019 (COVID-19) has led to major shifts in the management of colorectal cancer (CRC). This study aims to identify the impact and early outcomes of COVID-19 following CRC management at a tertiary referral center in Victoria, Australia. METHODS This was a retrospective study, utilizing the Australian Comprehensive Cancer Outcomes and Research Database and inpatient records. Patients presenting for CRC management at our institution were identified coinciding with the first Victorian outbreak of COVID-19 (March 26 to September 26, 2020) (COVID). Management decisions including chemoradiotherapy utilization and surgical outcomes were analyzed within 6 months and compared with the corresponding period in 2019 (pre-COVID). RESULTS A total of 276 patients were included in this study (147 pre-COVID period, 129 COVID period). During the COVID period, more patients (47.6% vs. 60.5%; p = 0.033) presented symptomatically and less for surveillance (10.9% vs. 2.3%; p < 0.01). Eighty-four pre-COVID and 69 COVID period patients proceeded to surgery. The average time from diagnosis date to surgery was 15.6 days less during the COVID period. There were no significant differences in postoperative utilization of higher care (p = 0.74), complications (p = 0.93), median hospital length of stay (p = 0.67), 30-day readmission (p = 0.50), or 30-day reoperation (p = 0.74). In 1.6% of cases, pandemic impacts resulted in a change in management. CONCLUSION Presentation of patients with CRC varied, with a significant increase in symptomatic presentations and decreased numbers for surveillance. Through flexibility and change in practice, our institution helped improve access to surgical intervention and oncological therapies. Further prospective work is required to identify long-term outcomes and characterize the effects of ongoing disruptions.
Collapse
Affiliation(s)
- Christy Kei
- Department of Colorectal Surgery, Western Health, Melbourne, Australia
| | - Richard Gartrell
- Department of Surgery, Western Precinct, The University of Melbourne, Melbourne, Australia
| | - Yasser Arafat
- Department of Colorectal Surgery, Western Health, Melbourne, Australia
- Department of Surgery, Western Precinct, The University of Melbourne, Melbourne, Australia
| | - Elizabeth Degabriele
- Department of Surgery, Western Precinct, The University of Melbourne, Melbourne, Australia
| | - Josephine Yeung
- Department of Surgery, Western Precinct, The University of Melbourne, Melbourne, Australia
| | - Steven Chan
- Department of Surgery, Western Precinct, The University of Melbourne, Melbourne, Australia
| | - Ian Faragher
- Department of Colorectal Surgery, Western Health, Melbourne, Australia
| | - Justin M C Yeung
- Department of Colorectal Surgery, Western Health, Melbourne, Australia
- Department of Surgery, Western Precinct, The University of Melbourne, Melbourne, Australia
- Chronic Disease Alliance, Western Health, Melbourne, Australia
| |
Collapse
|
208
|
Wang H, Zhang Y, Zhang H, Cao H, Mao J, Chen X, Wang L, Zhang N, Luo P, Xue J, Qi X, Dong X, Liu G, Cheng Q. Liquid biopsy for human cancer: cancer screening, monitoring, and treatment. MedComm (Beijing) 2024; 5:e564. [PMID: 38807975 PMCID: PMC11130638 DOI: 10.1002/mco2.564] [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: 04/23/2023] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 05/30/2024] Open
Abstract
Currently, tumor treatment modalities such as immunotherapy and targeted therapy have more stringent requirements for obtaining tumor growth information and require more accurate and easy-to-operate tumor information detection methods. Compared with traditional tissue biopsy, liquid biopsy is a novel, minimally invasive, real-time detection tool for detecting information directly or indirectly released by tumors in human body fluids, which is more suitable for the requirements of new tumor treatment modalities. Liquid biopsy has not been widely used in clinical practice, and there are fewer reviews of related clinical applications. This review summarizes the clinical applications of liquid biopsy components (e.g., circulating tumor cells, circulating tumor DNA, extracellular vesicles, etc.) in tumorigenesis and progression. This includes the development process and detection techniques of liquid biopsies, early screening of tumors, tumor growth detection, and guiding therapeutic strategies (liquid biopsy-based personalized medicine and prediction of treatment response). Finally, the current challenges and future directions for clinical applications of liquid biopsy are proposed. In sum, this review will inspire more researchers to use liquid biopsy technology to promote the realization of individualized therapy, improve the efficacy of tumor therapy, and provide better therapeutic options for tumor patients.
Collapse
Affiliation(s)
- Hao Wang
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Yi Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Hao Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Hui Cao
- Department of PsychiatryThe School of Clinical Medicine, Hunan University of Chinese MedicineChangshaChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People’s Hospital of Hunan Province)ChangshaChina
| | - Jinning Mao
- Health Management CenterThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Xinxin Chen
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Liangchi Wang
- Department of NeurosurgeryFengdu People's Hospital, ChongqingChongqingChina
| | - Nan Zhang
- College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Peng Luo
- Department of OncologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Ji Xue
- Department of NeurosurgeryTraditional Chinese Medicine Hospital Dianjiang ChongqingChongqingChina
| | - Xiaoya Qi
- Health Management CenterThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Xiancheng Dong
- Department of Cerebrovascular DiseasesDazhou Central HospitalSichuanChina
| | - Guodong Liu
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Quan Cheng
- Department of NeurosurgeryXiangya Hospital, Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| |
Collapse
|
209
|
Harrold EC, Stadler ZK. Upper Gastrointestinal Cancers and the Role of Genetic Testing. Hematol Oncol Clin North Am 2024; 38:677-691. [PMID: 38458854 DOI: 10.1016/j.hoc.2024.01.006] [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: 03/10/2024]
Abstract
Beyond the few established hereditary cancer syndromes with an upper gastrointestinal cancer component, there is increasing recognition of the contribution of novel pathogenic germline variants (gPVs) to upper gastrointestinal carcinogenesis. The detection of gPVs has potential implications for novel treatment approaches of the index cancer patient as well as long-term implications for surveillance and risk-reducing measures for cancer survivors and far-reaching implications for the patients' family. With widespread availability of multigene panel testing, new associations may be identified with germline-somatic integration being critical to determining true causality of novel gPVs. Comprehensive cancer care should incorporate both somatic and germline testing.
Collapse
Affiliation(s)
- Emily C Harrold
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland. https://twitter.com/EmilyHarrold6
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
210
|
Boukovala M, Westphalen CB, Probst V. Liquid biopsy into the clinics: Current evidence and future perspectives. THE JOURNAL OF LIQUID BIOPSY 2024; 4:100146. [PMID: 40027149 PMCID: PMC11863819 DOI: 10.1016/j.jlb.2024.100146] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 03/05/2025]
Abstract
As precision oncology has become a major part of the treatment landscape in oncology, liquid biopsies have developed as a particularly powerful tool as it surmounts several limitations of traditional tissue biopsies. These biopsies involve most commonly the isolation of circulating extracellular nucleic acids, including cell-free DNA (cfDNA) and circulating tumor DNA (ctDNA), as well as circulating tumor cells (CTCs), typically from blood. The clinical applications of liquid biopsies are diverse, encompassing the initial diagnosis and cancer detection, the application as a tool for prognostication in early and advanced tumor settings, the identification of potentially actionable alterations, the monitoring of response and resistance under systemic therapy and the detection of resistance mechanisms, the differentiation of distinct immune checkpoint blockade response patterns through serial samples, the prediction of immune checkpoint blockade responses based on initial liquid biopsy characteristics and the assessment of tumor heterogeneity. Moreover, molecular relapse monitoring in early-stage cancers and the personalization of adjuvant or additive therapy via MRD have become a major field of research in recent years. Compared to tissue biopsies, liquid biopsies are less invasive and can be collected serially, offering real-time molecular insights. Furthermore, liquid biopsies may allow for a more holistic evaluation of a patient's disease, as they assess material from all tumor sites and can theoretically reflect tumor heterogeneity. Furthermore, quicker turnaround-time also constitutes an advantage of liquid biopsies. Disadvantages or hurdles include the challenge of detecting low amounts of tumor deposits in peripheral blood or other fluids and the potential of different amounts tumor-shedding from different metastatic sites, as well as potentially false-positive from clonal hematopoietic mutations of indeterminate potential (CHIP) mutations. The clinical utility of liquid biopsies still must be validated in most settings and further research has to be done. Clinal trials including alternate bodily fluids and leveraging AI-technology are expected to revolutionize the field of liquid biopsies.
Collapse
|
211
|
Shroff RT, Bachini M. Treatment options for biliary tract cancer: unmet needs, new targets and opportunities from both physicians' and patients' perspectives. Future Oncol 2024; 20:1435-1450. [PMID: 38861288 PMCID: PMC11376410 DOI: 10.1080/14796694.2024.2340959] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 04/05/2024] [Indexed: 06/12/2024] Open
Abstract
Biliary tract cancer (BTC) is a rare cancer with poor prognosis, characterized by considerable pathophysiological and molecular heterogeneity. While this makes it difficult to treat, it also provides targeted therapy opportunities. Current standard-of-care is chemotherapy ± immunotherapy, but several targeted agents have recently been approved. The current investigational landscape in BTC emphasizes the importance of biomarker testing at diagnosis. MDM2/MDMX are important negative regulators of the tumor suppressor p53 and provide an additional target in BTC (∼5-8% of tumors are MDM2-amplified). Brigimadlin (BI 907828) is a highly potent MDM2-p53 antagonist that has shown antitumor activity in preclinical studies and promising results in early clinical trials; enrollment is ongoing in a potential registrational trial for patients with BTC.
Collapse
Affiliation(s)
- Rachna T Shroff
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85719, USA
| | - Melinda Bachini
- Cholangiocarcinoma Foundation, 5526 West 13400 South, #510, Herriman, UT USA
| |
Collapse
|
212
|
Raffaini G, Elli S, Catauro M, D’Angelo A. Different Drug Mobilities in Hydrophobic Cavities of Host-Guest Complexes between β-Cyclodextrin and 5-Fluorouracil at Different Stoichiometries: A Molecular Dynamics Study in Water. Int J Mol Sci 2024; 25:5888. [PMID: 38892075 PMCID: PMC11172661 DOI: 10.3390/ijms25115888] [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: 04/27/2024] [Revised: 05/11/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Cyclodextrins (CDs) are cyclic oligosaccharides able to form noncovalent water-soluble complexes useful in many different applications for the solubilization, delivery, and greater bioavailability of hydrophobic drugs. The complexation of 5-fluorouracil (5-FU) with natural or synthetic cyclodextrins permits the solubilization of this poorly soluble anticancer drug. In this theoretical work, the complexes between β-CD and 5-FU are investigated using molecular mechanics (MM) and molecular dynamics (MD) simulations in water. The inclusion complexes are formed thanks to the favorable intermolecular interactions between β-CD and 5-FU. Both 1:1 and 1:2 β-CD/5-FU stoichiometries are investigated, providing insight into their interaction geometries and stability over time in water. In the 1:2 β-CD/5-FU complexes, the intermolecular interactions affect the drug's mobility, suggesting a two-step release mechanism: a fast release for the more exposed and hydrated drug molecule, with greater freedom of movement near the β-CD rims, and a slow one for the less-hydrated and well-encapsulated and confined drug. MD simulations study the intermolecular interactions between drugs and specific carriers at the atomistic level, suggesting a possible release mechanism and highlighting the role of the impact of the drug concentration on the kinetics process in water. A comparison with experimental data in the literature provides further insights.
Collapse
Affiliation(s)
- Giuseppina Raffaini
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza L. Da Vinci 32, 20131 Milano, Italy
- INSTM, National Consortium of Materials Science and Technology, Local Unit Politecnico di Milano, 50121 Milano, Italy
| | - Stefano Elli
- Istituto di Ricerche Chimiche e Biochimiche ‘G. Ronzoni’, Via Giuseppe Colombo 81, 20133 Milano, Italy;
| | - Michelina Catauro
- Department of Engineering, University of Campania “Luigi Vanvitelli”, Via Roma 29, 81031 Aversa, Italy; (M.C.); (A.D.)
| | - Antonio D’Angelo
- Department of Engineering, University of Campania “Luigi Vanvitelli”, Via Roma 29, 81031 Aversa, Italy; (M.C.); (A.D.)
| |
Collapse
|
213
|
Mehdi M, Szabo A, Shreenivas A, Thomas JP, Tsai S, Christians KK, Evans DB, Clarke CN, Hall WA, Erickson B, Ahmed G, Thapa B, McFall T, George B, Kurzrock R, Kamgar M. Chemotherapy-free treatment targeting fusions and driver mutations in KRAS wild-type pancreatic ductal adenocarcinoma, a case series. Ther Adv Med Oncol 2024; 16:17588359241253113. [PMID: 38770091 PMCID: PMC11104030 DOI: 10.1177/17588359241253113] [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: 12/04/2023] [Accepted: 04/18/2024] [Indexed: 05/22/2024] Open
Abstract
Background KRAS wild-type (WT) pancreatic ductal adenocarcinoma (PDAC) represents a distinct entity with unique biology. The therapeutic impact of matched targeted therapy in these patients in a real-world setting, to date, is less established. Objectives The aim of our study was to review our institutional database to identify the prevalence of actionable genomic alterations in patients with KRAS-WT tumors and to evaluate the therapeutic impact of matched targeted therapy in these patients. Design We reviewed electronic medical records of patients with KRAS-WT PDAC and advanced disease (n = 14) who underwent clinical-grade tissue ± liquid next-generation sequencing (315-648 genes for tissue) between years 2015 and 2021. Methods Demographic and disease characteristics were summarized using descriptive parameters. Progression-free survival (PFS) and overall survival (OS) were estimated using the Kaplan-Meier method. Results Of 236 PDAC patients, 14 had advanced/metastatic disease with KRAS-WT tumors. Median age at diagnosis was 66 years. There was a high frequency of potentially actionable genomic alterations, including three (21%) with BRAF alterations, two (14%) with fusions [RET-PCM1 and FGFR2-POC1B (N = 1 each)]; and one with a druggable EGFR (EGFR E746_A755delISERD) variant; two other patients had an STK11 and a MUTYH alteration. Five patients were treated with matched targeted therapy, with three having durable benefit: (i) erlotinib for EGFR-altered tumor, followed by osimertinib/capmatinib when MET amplification emerged (first-line therapy); (ii) pralsetinib for RET fusion (fifth line); and (iii) dabrafenib/trametinib for BRAF N486_P490del (third line). Duration of time on chemotherapy-free matched targeted therapy for these patients was 17+, 11, and 18+ months, respectively. Conclusion Sustained therapeutic benefit can be achieved in a real-world setting in a subset of patients with advanced/metastatic KRAS-WT PDAC treated with chemotherapy-free matched targeted agents. Prospective studies are warranted.
Collapse
Affiliation(s)
- Maahum Mehdi
- Department of Medicine, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Aniko Szabo
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Aditya Shreenivas
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - James P. Thomas
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Susan Tsai
- Department of Surgery, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Kathleen K. Christians
- Department of Surgery, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Douglas B. Evans
- Department of Surgery, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Callisia N. Clarke
- Department of Surgery, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - William A. Hall
- Department of Radiation Oncology, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Beth Erickson
- Department of Radiation Oncology, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Gulrayz Ahmed
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Bicky Thapa
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Thomas McFall
- Department of Biochemistry, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Ben George
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Razelle Kurzrock
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| | - Mandana Kamgar
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin and the LaBahn Pancreatic Cancer Program, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA
| |
Collapse
|
214
|
Chen Z, Zhou J, Chen W, Wu T, Lian K, Shen T. Neoadjuvant envafolimab in a patient with MSI-H/dMMR colon cancer: a case report and literature review. Immunotherapy 2024; 16:649-657. [PMID: 39259508 PMCID: PMC11404695 DOI: 10.1080/1750743x.2024.2350355] [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: 01/15/2024] [Accepted: 04/26/2024] [Indexed: 09/13/2024] Open
Abstract
Clinical evidences of neoadjuvant immunotherapy in patients with mismatch repair deficient/microsatellite instability-high status (dMMR/MSI-H) colorectal cancer have not been well received. A 36-year-old man complained of recurrent right upper quadrant pain for more than 1 year, and the symptoms were not significantly relieved after 10 days of oral Changyanning tablet. The patient was finally diagnosed as dMMR/MSI-H colon cancer. Tumor regression was achieved after seven cycles of envafolimab treatment, and the patient obtained postoperative pathological complete response (pCR). Here, we report a case of MSI-H/dMMR transverse colon cancer, who obtained pCR after neoadjuvant envafolimab (a novel subcutaneous single-domain anti-PD-L1 antibody) with a favorable safety profile, aiming to enhance the experiences of comprehensive diagnosis and treatment of colon cancer.
Collapse
Affiliation(s)
- Ziwei Chen
- Department of Colorectal Surgery, The Third Affiliated Hospital of Kunming Medical University/Yunnan Tumor Hospital, Kunming, China
| | - Jingrui Zhou
- Department of Colorectal Surgery, The Third Affiliated Hospital of Kunming Medical University/Yunnan Tumor Hospital, Kunming, China
| | - Weimin Chen
- Department of Colorectal Surgery, The Third Affiliated Hospital of Kunming Medical University/Yunnan Tumor Hospital, Kunming, China
| | - Tao Wu
- Department of Colorectal Surgery, The Third Affiliated Hospital of Kunming Medical University/Yunnan Tumor Hospital, Kunming, China
| | - Ke Lian
- Department of Colorectal Surgery, The Third Affiliated Hospital of Kunming Medical University/Yunnan Tumor Hospital, Kunming, China
| | - Tao Shen
- Department of Colorectal Surgery, The Third Affiliated Hospital of Kunming Medical University/Yunnan Tumor Hospital, Kunming, China
| |
Collapse
|
215
|
Martínez-Galán J, Jiménez-Luna C, Rodriguez I, Maza E, García-Collado C, Rodríguez-Fernández A, López-Hidalgo JL, Caba O. Metastatic pancreatic and lung cancer patient in complete remission following immunotherapy: A case report and review of literature. World J Gastrointest Oncol 2024; 16:2233-2240. [PMID: 38764840 PMCID: PMC11099454 DOI: 10.4251/wjgo.v16.i5.2233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/09/2024] [Accepted: 03/13/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND Metastatic pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with dispiriting survival data. Immunotherapy is a promising approach to many cancer types, but achieves poor outcomes in advanced PDAC due to its immunosuppressive tumor microenvironment. We describe a case of metastatic PDAC effectively treated with pembrolizumab. CASE SUMMARY We report the case of a 67-year-old woman with unresectable locally advanced PDAC, treated with gemcitabine plus nab-paclitaxel followed by radiotherapy plus capecitabine. At nine months, pancreatic tumor progression was observed at the level of the hepatic hilum with the appearance of a new pulmonary nodule suggestive of a second primary, confirmed by left lung biopsy. Systemic immunotherapy was then initiated with pembrolizumab, an immune checkpoint inhibitor targeting programmed cell death protein-1 that covers the two tumor types. The patient showed a complete metabolic response that was maintained throughout the treatment. The patient continues to be disease-free at 5.6 years since the start of immunotherapy. CONCLUSION These results suggest that the administration of pembrolizumab after chemoradiotherapy has a beneficial effect in patients with metastatic PDAC. To our knowledge, this is the first reported case of a patient with metastatic PDAC and metastatic lung cancer showing such a long-lasting complete response after pembrolizumab treatment without curative surgery. Further studies are required to determine biomarkers that identify PDAC patients most likely to benefit from this immunotherapy.
Collapse
Affiliation(s)
- Joaquina Martínez-Galán
- Department of Medical Oncology, Virgen de las Nieves University Hospital, Granada 18014, Spain
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
| | - Cristina Jiménez-Luna
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada 18016, Spain
| | - Isabel Rodriguez
- Department of Medical Oncology, Virgen de las Nieves University Hospital, Granada 18014, Spain
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
| | - Elisabeth Maza
- Department of Medical Oncology, Virgen de las Nieves University Hospital, Granada 18014, Spain
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
| | - Carlos García-Collado
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
- Pharmacogenetics Unit, Virgen de las Nieves University Hospital, Granada 18014, Spain
| | - Antonio Rodríguez-Fernández
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
- Department of Medicina Nuclear, Virgen de las Nieves University Hospital, Granada 18014, Spain
| | - Javier Luis López-Hidalgo
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
- Department of Anatomía Patológica, Virgen de las Nieves University Hospital, Granada 18014, Spain
| | - Octavio Caba
- Instituto de Investigación Biosanitaria, Ibs. Granada, Granada 18012, Spain
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada 18016, Spain
| |
Collapse
|
216
|
Cronin M, Lowery A, Kerin M, Wijns W, Soliman O. Risk Prediction, Diagnosis and Management of a Breast Cancer Patient with Treatment-Related Cardiovascular Toxicity: An Essential Overview. Cancers (Basel) 2024; 16:1845. [PMID: 38791923 PMCID: PMC11120055 DOI: 10.3390/cancers16101845] [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: 04/12/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Breast cancer is amongst the most common invasive cancers in adults. There are established relationships between anti-cancer treatments for breast cancer and cardiovascular side effects. In recent years, novel anti-cancer treatments have been established, as well as the availability of multi-modal cardiac imaging and the sophistication of treatment for cardiac disease. This review provides an in-depth overview regarding the interface of breast cancer and cancer therapy-related cardiovascular toxicity. Specifically, it reviews the pathophysiology of breast cancer, the method of action in therapy-related cardiovascular toxicity from anti-cancer treatment, the use of echocardiography, cardiac CT, MRI, or nuclear medicine as diagnostics, and the current evidence-based treatments available. It is intended to be an all-encompassing review for clinicians caring for patients in this situation.
Collapse
Affiliation(s)
- Michael Cronin
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland
| | - Aoife Lowery
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
| | - Michael Kerin
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- Discipline of Surgery, Lambe Institute for Translational Research, University of Galway, H91 V4AY Galway, Ireland
| | - William Wijns
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
| | - Osama Soliman
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
- Discipline of Surgery, Lambe Institute for Translational Research, University of Galway, H91 V4AY Galway, Ireland
| |
Collapse
|
217
|
Mondal D, Shinde S, Sinha V, Dixit V, Paul S, Gupta RK, Thakur S, Vishvakarma NK, Shukla D. Prospects of liquid biopsy in the prognosis and clinical management of gastrointestinal cancers. Front Mol Biosci 2024; 11:1385238. [PMID: 38770216 PMCID: PMC11103528 DOI: 10.3389/fmolb.2024.1385238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/08/2024] [Indexed: 05/22/2024] Open
Abstract
Gastrointestinal (GI) cancers account for one-fourth of the global cancer incidence and are incriminated to cause one-third of cancer-related deaths. GI cancer includes esophageal, gastric, liver, pancreatic, and colorectal cancers, mostly diagnosed at advanced stages due to a lack of accurate markers for early stages. The invasiveness of diagnostic methods like colonoscopy for solid biopsy reduces patient compliance as it cannot be frequently used to screen patients. Therefore, minimally invasive approaches like liquid biopsy may be explored for screening and early identification of gastrointestinal cancers. Liquid biopsy involves the qualitative and quantitative determination of certain cancer-specific biomarkers in body fluids such as blood, serum, saliva, and urine to predict disease progression, therapeutic tolerance, toxicities, and recurrence by evaluating minimal residual disease and its correlation with other clinical features. In this review, we deliberate upon various tumor-specific cellular and molecular entities such as circulating tumor cells (CTCs), tumor-educated platelets (TEPs), circulating tumor DNA (ctDNA), cell-free DNA (cfDNA), exosomes, and exosome-derived biomolecules and cite recent advances pertaining to their use in predicting disease progression, therapy response, or risk of relapse. We also discuss the technical challenges associated with translating liquid biopsy into clinical settings for various clinical applications in gastrointestinal cancers.
Collapse
Affiliation(s)
- Deepankar Mondal
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
| | - Sapnita Shinde
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
| | - Vibha Sinha
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
| | - Vineeta Dixit
- Department of Botany, Sri Sadguru Jagjit Singh Namdhari College, Garhwa, Jharkhand, India
| | - Souvik Paul
- Department of Surgical Gastroenterology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| | - Rakesh Kumar Gupta
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| | | | | | - Dhananjay Shukla
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
| |
Collapse
|
218
|
Kinos S, Hagman H, Halonen P, Soveri LM, O'Reilly M, Pfeiffer P, Frödin JE, Sorbye H, Heervä E, Liposits G, Kallio R, Ålgars A, Ristamäki R, Salminen T, Bärlund M, Shah CH, McDermott R, Röckert R, Flygare P, Kwakman J, Teske A, Punt C, Glimelius B, Österlund P. Detailed analysis of metastatic colorectal cancer patients who developed cardiotoxicity on another fluoropyrimidine and switched to S-1 treatment (subgroup analysis of the CardioSwitch-study). Acta Oncol 2024; 63:248-258. [PMID: 38698698 PMCID: PMC11332541 DOI: 10.2340/1651-226x.2024.24023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/16/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND AND PURPOSE The CardioSwitch-study demonstrated that patients with solid tumors who develop cardiotoxicity on capecitabine or 5-fluorouracil (5-FU) treatment can be safely switched to S-1, an alternative fluoropyrimidine (FP). In light of the European Medicines Agency approval of S-1 in metastatic colorectal cancer (mCRC), this analysis provides more detailed safety and efficacy information, and data regarding metastasectomy and/or local ablative therapy (LAT), on the mCRC patients from the original study. MATERIALS AND METHODS This retrospective cohort study was conducted at 12 European centers. The primary endpoint was recurrence of cardiotoxicity after switch. For this analysis, safety data are reported for 78 mCRC patients from the CardioSwitch cohort (N = 200). Detailed efficacy and outcomes data were available for 66 mCRC patients. RESULTS Data for the safety of S-1 in mCRC patients were similar to the original CardioSwitch cohort and that expected for FP-based treatment, with no new concerns. Recurrent cardiotoxicity (all grade 1) with S-1-based treatment occurred in 4/78 (5%) mCRC patients; all were able to complete FP treatment. Median progression-free survival from initiation of S-1-based treatment was 9.0 months and median overall survival 26.7 months. Metastasectomy and/or LAT was performed in 33/66 (50%) patients, and S-1 was successfully used in recommended neoadjuvant/conversion or adjuvant-like combination regimens and schedules as for standard FPs. INTERPRETATION S-1 is a safe and effective FP alternative when mCRC patients are forced to discontinue 5-FU or capecitabine due to cardiotoxicity and can be safely used in the standard recommended regimens, settings, and schedules.
Collapse
Affiliation(s)
- Sampsa Kinos
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland; Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Helga Hagman
- Department of Oncology, Skåne University Hospital, Malmö, Sweden
| | - Päivi Halonen
- Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Leena-Maija Soveri
- Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mary O'Reilly
- Department of Oncology, St Vincent's University Hospital and University College Dublin, Dublin, Ireland
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Jan-Erik Frödin
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Eetu Heervä
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Gabor Liposits
- Department of Oncology, Regional Hospital West Jutland, Hjørring, Denmark
| | - Raija Kallio
- Department of Oncology, Oulu University and University Hospital, Oulu, Finland
| | - Annika Ålgars
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Raija Ristamäki
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Tapio Salminen
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland; Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Maarit Bärlund
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland; Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Carl-Henrik Shah
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Ray McDermott
- Department of Oncology, St Vincent's University Hospital and University College Dublin, Dublin, Ireland
| | | | - Petra Flygare
- Department of Oncology, Sundsvall Hospital, Sundsvall, Sweden
| | - Johannes Kwakman
- Department of Medical Oncology, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Arco Teske
- Department of Cardiology, University Medical Centre, Utrecht University, Utrecht, The Netherlands
| | - Cornelis Punt
- Depatment of Epidemiology, Jules Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherland
| | | | - Pia Österlund
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland; Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland; Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden; rTema Cancer, Department of GI-cancer, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
219
|
Agnihotri A, Ramasubbu SK, Bandyopadhyay A, Bidarolli M, Nath UK, Das B. Prevalence, Attributes, and Risk Factors of QT-Interval-Prolonging Drugs and Potential Drug-Drug Interactions in Cancer Patients: A Prospective Study in a Tertiary Care Hospital. Cureus 2024; 16:e60492. [PMID: 38882995 PMCID: PMC11180424 DOI: 10.7759/cureus.60492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction Cancer chemotherapy regimens include multiple classes of adjuvant drugs as supportive therapy. Because of the concurrent intake of other drugs (like antiemetics, antidepressants, analgesics, and antimicrobials), there is a heightened risk for possible QT interval prolongation. There is a dearth of evidence in the literature regarding the usage of QT-prolonging anticancer drugs and associated risk factors that have the propensity to prolong QT interval. The purpose was to explore the extent of the use of QT-interval-prolonging drugs and potential QT-prolonging drug-drug interactions (QT-DDIs) in cancer patients attending OPD in a tertiary-care hospital. Methods This was a hospital-based, cross-sectional, observational study. Risk stratification of QT-prolonging drugs for torsades de pointes (TdP) was done by the Arizona Center for Education and Research on Therapeutics (AzCERT)/CredibleMeds-lists, and potential QT-DDIs were determined with four online DDI-checker-software. Results In 1331 cancer patients, the overall prevalence of potential QT-prolonging drug utilization was 97.3%. Ondansetron, pantoprazole, domperidone, and olanzapine were the most frequent QT-prolonging drugs in cancer patients. The top six antineoplastics with potential QT-prolonging and torsadogenic actions were capecitabine, oxaliplatin, imatinib, bortezomib, 5-fluorouracil, and bendamustine. Evidence-based pragmatic QTc interval prolongation risk assessment tools are imperative for cancer patients. Conclusion This study revealed a high prevalence of QT-prolonging drugs and QT-DDIs among cancer patients who are treated with anticancer and non-anticancer drugs. As a result, it's critical to take precautions, stay vigilant, and avoid QT-prolonging in clinical situations. Evidence-based pragmatic QTc interval prolongation risk assessment tools are needed for cancer patients.
Collapse
Affiliation(s)
- Akash Agnihotri
- Department of Pharmacology, Amrita School of Medicine, Faridabad, IND
| | - Saravana Kumar Ramasubbu
- Department of Pharmacology, Andaman and Nicobar Islands Institute of Medical Sciences, Port Blair, IND
| | - Arkapal Bandyopadhyay
- Department of Pharmacology, All India Institute of Medical Sciences, Kalyani, Kalyani, IND
| | - Manjunath Bidarolli
- Department of Pharmacology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| | - Uttam Kumar Nath
- Department of Medical Oncology and Hematology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| | - Biswadeep Das
- Department of Pharmacology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| |
Collapse
|
220
|
Zhang D, Dorman K, Westphalen CB, Haas M, Ormanns S, Neumann J, Seidensticker M, Ricke J, De Toni EN, Klauschen F, Algül H, Reisländer T, Boeck S, Heinemann V. Unresectable biliary tract cancer: Current and future systemic therapy. Eur J Cancer 2024; 203:114046. [PMID: 38626513 DOI: 10.1016/j.ejca.2024.114046] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/18/2024]
Abstract
For decades, treatment of advanced biliary tract cancer (BTC) was confined to the use of chemotherapy. In recent years however, the number of therapeutic options available for patients with unresectable BTC have drastically increased, with immunotherapy and targeted treatment gradually joining the ranks of guideline-recommended treatment regimens. The aim of the present review is to summarise the current knowledge on unresectable BTC focusing on epidemiology, anatomical distribution and current strategies for systemic treatment. We further outline ongoing clinical trials and provide an outlook on future therapeutic interventions. In the realm of gastrointestinal malignancies, the increasing number of systemic treatment options for BTC is finally delivering on the longstanding commitment to personalised oncology. This emphasises the need for considering a comprehensive genomic-based pathology assessment right from the initial diagnosis to fully leverage the expanding array of therapeutic options that have recently become accessible.
Collapse
Affiliation(s)
- Danmei Zhang
- Department of Medicine III, LMU University Hospital, LMU Munich and Comprehensive Cancer Center Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Klara Dorman
- Department of Medicine III, LMU University Hospital, LMU Munich and Comprehensive Cancer Center Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - C Benedikt Westphalen
- Department of Medicine III, LMU University Hospital, LMU Munich and Comprehensive Cancer Center Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Michael Haas
- Department of Medicine III, LMU University Hospital, LMU Munich and Comprehensive Cancer Center Munich, Marchioninistr. 15, 81377 Munich, Germany; Department of Hematology and Oncology, München Klinik Neuperlach, Munich, Germany
| | - Steffen Ormanns
- Institute of Pathology, Faculty of Medicine, LMU Munich, Germany; Innpath GmbH, Tirolkliniken, Innsbruck, Austria
| | - Jens Neumann
- Institute of Pathology, Faculty of Medicine, LMU Munich, Germany
| | - Max Seidensticker
- Department of Radiology, LMU University Hospital, LMU Munich, Germany
| | - Jens Ricke
- Department of Radiology, LMU University Hospital, LMU Munich, Germany
| | - Enrico N De Toni
- Department of Medicine II, LMU University Hospital, LMU Munich, Germany; Boehringer Ingelheim, Clinical Program Lead, Bingerstrasse 137, Ingelheim am Rhein 55218, Germany
| | | | - Hana Algül
- Comprehensive Cancer Center Munich TUM, Institute for Tumor Metabolism, Technical University of Munich, Munich, Germany
| | - Timo Reisländer
- SERVIER Deutschland GmbH, Medical Affairs, Elsenheimerstr. 53, 80687 Munich, Germany
| | - Stefan Boeck
- Department of Medicine III, LMU University Hospital, LMU Munich and Comprehensive Cancer Center Munich, Marchioninistr. 15, 81377 Munich, Germany; Department of Hematology and Oncology, München Klinik Neuperlach, Munich, Germany
| | - Volker Heinemann
- Department of Medicine III, LMU University Hospital, LMU Munich and Comprehensive Cancer Center Munich, Marchioninistr. 15, 81377 Munich, Germany.
| |
Collapse
|
221
|
Oh Y, Yoon SM, Lee J, Park JH, Lee S, Hong T, Chung LI, Sudhaman S, Riddell T, Palsuledesai CC, Krainock M, Liu MC, Chae YK. Personalized, tumor-informed, circulating tumor DNA assay for detecting minimal residual disease in non-small cell lung cancer patients receiving curative treatments. Thorac Cancer 2024; 15:1095-1102. [PMID: 38558374 PMCID: PMC11062881 DOI: 10.1111/1759-7714.15281] [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: 01/07/2024] [Revised: 02/24/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) has emerged as a prognostic and predictive biomarker for detection of minimal residual disease (MRD), monitoring treatment response, and early detection of recurrence in cancer patients. In this study, we explored the utility of ctDNA-based MRD detection to predict recurrence in a real-world cohort of primarily early-stage non-small cell lung cancer (NSCLC) patients treated with curative intent. METHODS Longitudinal plasma samples were collected post curative-intent treatment from 36 patients with stage I-IV NSCLC. A personalized, tumor-informed assay was used to detect and quantify ctDNA in plasma samples. RESULTS Of the 24 patients with plasma samples available during the MRD window (within 6 months of curative surgery and before adjuvant therapy), ctDNA was detectable in two patients. Patients with ctDNA-positivity during the MRD window were 15 times more likely to recur compared to ctDNA-negative patients (HR: 15.0, 95% CI: 1.0-253.0, p = 0.010). At any time post-curative intent treatment, ctDNA-positivity was associated with significantly poorer recurrence-free survival compared to persistently ctDNA-negative patients (p < 0.0001). CONCLUSION Our real-world data indicate that longitudinal, personalized, tumor-informed ctDNA monitoring is a valuable tool in patients with NSCLC receiving curative treatment to identify patients at high risk for recurrence.
Collapse
Affiliation(s)
- Youjin Oh
- Feinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- Department of internal medicineJohn H. Stroger Hospital of Cook CountyChicagoIllinoisUSA
| | - Sung Mi Yoon
- Feinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- North Central Bronx Hospital, Albert Einstein College of MedicineBronxNew YorkUSA
| | - Jeeyeon Lee
- Feinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- Kyungpook National University School of Medicine, Kyungpook National University Chilgok HospitalDaeguRepublic of Korea
| | - Joo Hee Park
- Feinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Soowon Lee
- Feinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- Baylor UniversityWacoTexasUSA
| | - Timothy Hong
- Feinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | | | | | | | | | | | | | - Young Kwang Chae
- Feinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
222
|
Moffat GT, Hu ZI, Meric-Bernstam F, Kong EK, Pavlick D, Ross JS, Murugesan K, Kwong L, De Armas AD, Korkut A, Javle M, Knox JJ. KRAS Allelic Variants in Biliary Tract Cancers. JAMA Netw Open 2024; 7:e249840. [PMID: 38709532 PMCID: PMC11074811 DOI: 10.1001/jamanetworkopen.2024.9840] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/06/2024] [Indexed: 05/07/2024] Open
Abstract
Importance Biliary tract cancers (BTCs) contain several actionable molecular alterations, including FGFR2, IDH1, ERBB2 (formerly HER2), and KRAS. KRAS allelic variants are found in 20% to 30% of BTCs, and multiple KRAS inhibitors are currently under clinical investigation. Objectives To describe the genomic landscape, co-sequence variations, immunophenotype, genomic ancestry, and survival outcomes of KRAS-mutated BTCs and to calculate the median overall survival (mOS) for the most common allelic variants. Design, Setting, and Participants This retrospective, multicenter, pooled cohort study obtained clinical and next-generation sequencing data from multiple databases between January 1, 2017, and December 31, 2022. These databases included Princess Margaret Cancer Centre, MD Anderson Cancer Center, Foundation Medicine, American Association for Cancer Research Project GENIE, and cBioPortal for Cancer Genomics. The cohort comprised patients with BTCs who underwent genomic testing. Main Outcome and Measure The main outcome was mOS, defined as date of diagnosis to date of death, which was measured in months. Results A total of 7457 patients (n = 3773 males [50.6%]; mean [SD] age, 63 [5] years) with BTCs and genomic testing were included. Of these patients, 5813 had clinical outcome data available, in whom 1000 KRAS-mutated BTCs were identified. KRAS allelic variants were highly prevalent in perihilar cholangiocarcinoma (28.6%) and extrahepatic cholangiocarcinoma (36.1%). Thirty-six KRAS allelic variants were identified, and the prevalence rates in descending order were G12D (41%), G12V (23%), and Q61H (8%). The variant G12D had the highest mOS of 25.1 (95% CI, 22.0-33.0) months compared with 22.8 (95% CI, 19.6-31.4) months for Q61H and 17.8 (95% CI, 16.3-23.1) months for G12V variants. The majority of KRAS-mutated BTCs (98.9%) were not microsatellite instability-high and had low tumor mutational burden (ranging from a median [IQR] of 1.2 (1.2-2.5) to a mean [SD] of 3.3 [1.3]). Immune profiling through RNA sequencing of KRAS and NRAS-mutated samples showed a pattern toward a more immune-inflamed microenvironment with higher M1 macrophage activation (0.16 vs 0.12; P = .047) and interferon-γ expression compared with wild-type tumors. The G12D variant remained the most common KRAS allelic variant in all patient ancestries. Patients with admixed American ancestry had the highest proportion of G12D variant (45.0%). Conclusions and Relevance This cohort study found that KRAS allelic variants were relatively common and may be potentially actionable genomic alterations in patients with BTCs, especially perihilar cholangiocarcinoma and extrahepatic cholangiocarcinoma. The findings add to the growing data on genomic and immune landscapes of KRAS allelic variants in BTCs and are potentially of value to the planning of specific therapies for this heterogeneous patient group.
Collapse
Affiliation(s)
- Gordon Taylor Moffat
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Zishuo Ian Hu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Funda Meric-Bernstam
- Department of Developmental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston
| | - Elisabeth Kathleen Kong
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston
| | - Dean Pavlick
- Foundation Medicine Inc, Cambridge, Massachusetts
| | - Jeffrey S. Ross
- Foundation Medicine Inc, Cambridge, Massachusetts
- State University of New York Upstate Medical University, Syracuse
| | | | - Lawrence Kwong
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Anaemy Danner De Armas
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston
| | - Milind Javle
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Jennifer J. Knox
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
223
|
Wang J, Song Z. Analysis of risk factors for postoperative recurrence of stage I colorectal cancer: a retrospective analysis of a large population. Front Surg 2024; 11:1388250. [PMID: 38712335 PMCID: PMC11072714 DOI: 10.3389/fsurg.2024.1388250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
Background Colorectal cancer (CRC) is the third most common cancer worldwide. Patients diagnosed with stage I CRC typically do not require postoperative adjuvant treatment. However, postoperative recurrence is present in at least 40% of patients with CRC and often occurs in those with stage I disease. This study aimed to elucidate the current status of recurrence and clinicopathological characteristics in patients with stage I CRC. Methods Data of indicated patients were obtained from 18 registries in Surveillance, Epidemiology, and End Results (SEER). The multivariable Fine-Gray regression model was used to identify the mortality risk of patients. Disparities in survival were analyzed using Kaplan-Meier curves. Logistic regression was employed to identify factors associated with recurrent risk overestimation. Results Our study indicated a recurrence rate of 15.04% (1,874/12,452) in stage I CRC cases. Notably, we identified race, age, T stage, and carcinoembryonic antigen (CEA) levels as independent risk factors for tumor recurrence, substantially impacting prognosis. Furthermore, gender, race (Black), age (>65 years), elevated CEA levels, and refusal or unknown status regarding radiotherapy significantly correlated with an adverse prognosis in patients with stage I CRC. Conclusions We identified certain key clinicopathological features of patients with stage I CRC and demonstrated the survival benefits of radiotherapy, offering a new perspective on stage I CRC follow-up and treatment recommendations.
Collapse
Affiliation(s)
- Jiawei Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
224
|
Imperial R, Mosalem O, Majeed U, Tran NH, Borad MJ, Babiker H. Second-Line Treatment of Pancreatic Adenocarcinoma: Shedding Light on New Opportunities and Key Talking Points from Clinical Trials. Clin Exp Gastroenterol 2024; 17:121-134. [PMID: 38650920 PMCID: PMC11034511 DOI: 10.2147/ceg.s390655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Despite improvements in overall cancer mortality, deaths related to pancreatic cancer continue to rise. Following first-line treatment, second-line options are significantly limited. Classically, first-line treatment consisted of either gemcitabine or 5-fluorouracil based systemic chemotherapy. Upon progression of disease or recurrence, subsequent second-line treatment is still gemcitabine or 5-fluorouracil based chemotherapy, depending on what was used in the first line and the timing of progression or recurrence. A better understanding of the molecular underpinnings of pancreatic adenocarcinoma (PDAC) has led to new treatment strategies including specifically targeting the desmoplastic stroma, cytokine signaling and actionable mutations. Furthermore, efforts are also directed to enhance the immunogenicity profile of PDAC's well-established immunologically "cold" tumor microenvironment. More recently, the outstanding response rates of chimeric antigen receptor T (CAR-T) cells in hematologic malignancies, have led to clinical trials to evaluate the treatment modality in PDAC. In this review, we summarize recently presented clinical trials for metastatic pancreatic adenocarcinoma with novel treatment approaches in the second line and beyond.
Collapse
Affiliation(s)
- Robin Imperial
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Osama Mosalem
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Umair Majeed
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Mitesh J Borad
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ, USA
| | - Hani Babiker
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
225
|
Zhao J, Yan D, Li Y, Xu X, Li F, Zhang S, Jin J, Qiu F. Simultaneous determination of 11 oral targeted antineoplastic drugs and 2 active metabolites by LC-MS/MS in human plasma and its application to therapeutic drug monitoring in cancer patients. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1237:124100. [PMID: 38547701 DOI: 10.1016/j.jchromb.2024.124100] [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: 01/23/2024] [Revised: 03/11/2024] [Accepted: 03/17/2024] [Indexed: 04/13/2024]
Abstract
Interindividual exposure differences have been identified in oral targeted antineoplastic drugs (OADs) owing to the pharmacogenetic background of the patients and their susceptibility to multiple factors, resulting in insufficient efficacy or adverse effects. Therapeutic drug monitoring (TDM) can prevent sub-optimal concentrations of OADs and improve their clinical treatment. This study aimed to develop and validate an LC-MS/MS method for the simultaneous quantification of 11 OADs (gefitinib, imatinib, lenvatinib, regorafenib, everolimus, osimertinib, sunitinib, tamoxifen, lapatinib, fruquintinib and sorafenib) and 2 active metabolites (N-desethyl sunitinib and Z-endoxifen) in human plasma. Protein precipitation was used to extract OADs from the plasma samples. Chromatographic separation was performed using an Eclipse XDB-C18 (4.6 × 150 mm, 5 μm) column with a gradient elution of the mobile phase composed of 2 mM ammonium acetate with 0.1 % formic acid in water (solvent A) and methanol (solvent B) at a flow rate of 0.8 mL/min. Mass analysis was performed using positive ion mode electrospray ionization in multiple-reaction monitoring mode. The developed method was validated following FDA bioanalytical guidelines. The calibration curves were linear over the range of 2-400 ng/mL for gefitinib, imatinib, lenvatinib, regorafenib, and everolimus; 1-200 ng/mL for osimertinib, sunitinib, N-desethyl sunitinib, tamoxifen, and Z-endoxifen; and 5-1000 ng/mL for lapatinib, fruquintinib, and sorafenib, with all coefficients of correlation above 0.99. The intra- and inter-day imprecision was below 12.81 %. This method was successfully applied to the routine TDM of gefitinib, lenvatinib, regorafenib, osimertinib, fruquintinib, and sorafenib to optimize the dosage regimens.
Collapse
Affiliation(s)
- Jing Zhao
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Dongming Yan
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Yue Li
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Xiaoqing Xu
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Fengling Li
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Shuang Zhang
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Jingyi Jin
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China.
| | - Furong Qiu
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China.
| |
Collapse
|
226
|
Wilbur HC, Le DT, Agarwal P. Immunotherapy of MSI Cancer: Facts and Hopes. Clin Cancer Res 2024; 30:1438-1447. [PMID: 38015720 DOI: 10.1158/1078-0432.ccr-21-1935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/14/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023]
Abstract
Microsatellite instability (MSI) is a tumor molecular phenotype that evolves from loss of function in the mismatch repair (MMR) proteins through deleterious germline mutations, epigenetic inactivation, or somatic biallelic mutations. This phenotype is characterized by genomic hyper-mutability, increased neoantigen expression, and a favorable, immune-rich tumor microenvironment. These features confer a greater likelihood of response to treatment with the class of agents known as immune checkpoint inhibitors (ICI) and, potentially, other immune-based therapeutics. MSI as a predictive biomarker for response to treatment with ICIs ultimately led to the first tissue-agnostic approval of pembrolizumab for advanced, previously treated MSI or deficient MMR (dMMR) tumors. Nevertheless, response to ICIs in dMMR/MSI tumors is not universal. Identifying predictors of response and elucidating mechanisms of immune escape will be crucial to continued successful treatment of this subset. In this review, we aim to describe the pathogenesis and key immunologic features of dMMR/MSI tumors, provide a brief overview of the currently approved treatments, and discuss promising novel immune-based therapeutics currently under investigation.
Collapse
Affiliation(s)
- H Catherine Wilbur
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Dung T Le
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Parul Agarwal
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
227
|
Jou E. Clinical and basic science aspects of innate lymphoid cells as novel immunotherapeutic targets in cancer treatment. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 209:1-60. [PMID: 39461748 DOI: 10.1016/bs.pmbts.2024.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Immunotherapy has revolutionised cancer treatment over the past decade, demonstrating remarkable efficacy across a broad range of cancer types. However, not all patients or cancer types respond to contemporary clinically-utilised immunotherapeutic strategies, which largely focus on harnessing adaptive immune T cells for cancer treatment. Accordingly, it is increasingly recognised that upstream innate immune pathways, which govern and orchestrate the downstream adaptive immune response, may prove critical in overcoming cancer immunotherapeutic resistance. Innate lymphoid cells (ILCs) are the most recently discovered major innate immune cell population. They have overarching roles in homeostasis and orchestrating protective immunity against pathogens. As innate immune counterparts of adaptive immune T cells, ILCs exert effector functions through the secretion of cytokines and direct cell-to-cell contact, with broad influence on the overall immune response. Importantly, dysregulation of ILC subsets have been associated with a range of diseases, including immunodeficiency disorders, allergy, autoimmunity, and more recently, cancer. ILCs may either promote or inhibit cancer initiation and progression depending on the cancer type and the specific ILC subsets involved. Critically, therapeutic targeting of ILCs and their associated cytokines shows promise against a wide range of cancer types in both preclinical models and early phase oncology clinical trials. This chapter provides a comprehensive overview of the current understanding of ILC subsets and the associated cytokines they produce in cancer pathogenesis, with specific focus on how these innate pathways are, or can be targeted, therapeutically to overcome therapeutic resistance and ultimately improve patient care.
Collapse
Affiliation(s)
- Eric Jou
- Department of Oncology, Oxford University Hospitals, University of Oxford, Oxford, United Kingdom; Kellogg College, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
228
|
Elez E, Cubillo A, Alfonso PG, Middleton MR, Chau I, Alkuzweny B, Alcasid A, Zhang X, Van Cutsem E. Binimetinib in combination with nivolumab or nivolumab and ipilimumab in patients with previously treated microsatellite-stable metastatic colorectal cancer with RAS mutations in an open-label phase 1b/2 study. BMC Cancer 2024; 24:446. [PMID: 38600471 PMCID: PMC11007903 DOI: 10.1186/s12885-024-12153-5] [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: 03/14/2023] [Accepted: 03/20/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND In patients with previously treated RAS-mutated microsatellite-stable (MSS) metastatic colorectal cancer (mCRC), a multicenter open-label phase 1b/2 trial was conducted to define the safety and efficacy of the MEK1/MEK2 inhibitor binimetinib in combination with the immune checkpoint inhibitor (ICI) nivolumab (anti-PD-1) or nivolumab and another ICI, ipilimumab (anti-CTLA4). METHODS In phase 1b, participants were randomly assigned to Arm 1A (binimetinib 45 mg twice daily [BID] plus nivolumab 480 mg once every 4 weeks [Q4W]) or Arm 1B (binimetinib 45 mg BID plus nivolumab 480 mg Q4W and ipilimumab 1 mg/kg once every 8 weeks [Q8W]) to determine the maximum tolerable dose (MTD) and recommended phase 2 dose (RP2D) of binimetinib. The MTD/RP2D was defined as the highest dosage combination that did not cause medically unacceptable dose-limiting toxicities in more than 35% of treated participants in Cycle 1. During phase 2, participants were randomly assigned to Arm 2A (binimetinib MTD/RP2D plus nivolumab) or Arm 2B (binimetinib MTD/RP2D plus nivolumab and ipilimumab) to assess the safety and clinical activity of these combinations. RESULTS In phase 1b, 21 participants were randomized to Arm 1A or Arm 1B; during phase 2, 54 participants were randomized to Arm 2A or Arm 2B. The binimetinib MTD/RP2D was determined to be 45 mg BID. In phase 2, no participants receiving binimetinib plus nivolumab achieved a response. Of the 27 participants receiving binimetinib, nivolumab, and ipilimumab, the overall response rate was 7.4% (90% CI: 1.3, 21.5). Out of 75 participants overall, 74 (98.7%) reported treatment-related adverse events (AEs), of whom 17 (22.7%) reported treatment-related serious AEs. CONCLUSIONS The RP2D binimetinib regimen had a safety profile similar to previous binimetinib studies or nivolumab and ipilimumab combination studies. There was a lack of clinical benefit with either drug combination. Therefore, these data do not support further development of binimetinib in combination with nivolumab or nivolumab and ipilimumab in RAS-mutated MSS mCRC. TRIAL REGISTRATION NCT03271047 (09/01/2017).
Collapse
Affiliation(s)
- Elena Elez
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Antonio Cubillo
- Centro Integral, Oncológico Clara Campal, HM CIOCC, Madrid, Spain
- Facultad HM Hospitales de Ciencias de La Salud UCJC, 28050, Madrid, Spain
| | - Pilar Garcia Alfonso
- Medical Oncology Service, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, Madrid, Spain
| | - Mark R Middleton
- Department of Oncology, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Ian Chau
- Gastrointestinal Unit, Royal Marsden Hospital, London & Surrey, UK
| | | | | | | | - Eric Van Cutsem
- University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium
| |
Collapse
|
229
|
Niger M, Nichetti F, Fornaro L, Pircher C, Morano F, Palermo F, Rimassa L, Pressiani T, Berardi R, Gardini AC, Sperti E, Salvatore L, Melisi D, Bergamo F, Siena S, Mosconi S, Longarini R, Arcangeli G, Corallo S, Delliponti L, Tamberi S, Fea E, Brandi G, Rapposelli IG, Salati M, Baili P, Miceli R, Ljevar S, Cavallo I, Sottotetti E, Martinetti A, Busset MDD, Sposito C, Di Bartolomeo M, Pietrantonio F, de Braud F, Mazzaferro V. A phase II/III randomized clinical trial of CisPlatin plUs Gemcitabine and Nabpaclitaxel (GAP) as pReoperative chemotherapy versus immediate resection in patIents with resecTable BiliarY Tract Cancers (BTC) at high risk for recurrence: PURITY study. BMC Cancer 2024; 24:436. [PMID: 38589856 PMCID: PMC11003088 DOI: 10.1186/s12885-024-12225-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/02/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Biliary tract cancers (BTCs) are rare and lethal cancers, with a 5-year survival inferior to 20%(1-3). The only potential curative treatment is surgical resection. However, despite complex surgical procedures that have a remarkable risk of postoperative morbidity and mortality, the 5-year survival rate after radical surgery (R0) is 20-40% and recurrence rates are up to ~ 75%(4-6). Up to ~ 40% of patients relapse within 12 months after resection, and half of these patient will recur systemically(4-6). There is no standard of care for neoadjuvant chemotherapy (NAC) in resectable BTC, but retrospective reports suggest its potential benefit (7, 8). METHODS PURITY is a no-profit, multicentre, randomized phase II/III trial aimed at evaluating the efficacy of the combination of gemcitabine, cisplatin and nabpaclitaxel (GAP) as neoadjuvant treatment in patients with resectable BTC at high risk for recurrence. Primary objective of this study is to evaluate the efficacy of neoadjuvant GAP followed by surgery as compared to upfront surgery, in terms of 12-month progression-free survival for the phase II part and of progression free survival (PFS) for the phase III study. Key Secondary objectives are event free survival (EFS), relapse-free survival, (RFS), overall survival (OS), R0/R1/R2 resection rate, quality of life (QoL), overall response rate (ORR), resectability. Safety analyses will include toxicity rate and perioperative morbidity and mortality rate. Exploratory studies including Next-Generation Sequencing (NGS) in archival tumor tissues and longitudinal ctDNA analysis are planned to identify potential biomarkers of primary resistance and prognosis. DISCUSSION Considering the poor prognosis of resected BTC experiencing early tumor recurrence and the negative prognostic impact of R1/R2 resections, PURITY study is based on the rationale that NAC may improve R0 resection rates and ultimately patients' outcomes. Furthermore, NAC should allow early eradication of microscopic distant metastases, undetectable by imaging but already present at the time of diagnosis and avoid mortality and morbidity associated with resection for patients with rapid progression or worsening general condition during neoadjuvant therapy. The randomized PURITY study will evaluate whether patients affected by BTC at high risk from recurrence benefit from a neoadjuvant therapy with GAP regimen as compared to immediate surgery. TRIAL REGISTRATION PURITY is registered at ClinicalTrials.gov (NCT06037980) and EuCT(2023-503295-25-00).
Collapse
Affiliation(s)
- Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian, 1, 20133, Milan, Italy.
| | - Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian, 1, 20133, Milan, Italy
- Computational Oncology, Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lorenzo Fornaro
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Chiara Pircher
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian, 1, 20133, Milan, Italy
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian, 1, 20133, Milan, Italy
| | - Federica Palermo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian, 1, 20133, Milan, Italy
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Tiziana Pressiani
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Rossana Berardi
- Clinica Di Oncologia Medica, A.O.U. Delle Marche, Università Politecnica Delle Marche, Ancona, Italy
| | - Andrea Casadei Gardini
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Medical Oncology, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Elisa Sperti
- Department of Oncology, University of Turin, AO Ordine Mauriziano Hospital, Turin, Italy
| | - Lisa Salvatore
- Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Oncologia Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Davide Melisi
- Digestive Molecular Clinical Oncology Research Unit, Università Degli Studi Di Verona, Verona, Italy
- Investigational Cancer Therapeutics Clinical Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Francesca Bergamo
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Salvatore Siena
- Department of Hematology Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | | | | | - Salvatore Corallo
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Laura Delliponti
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Stefano Tamberi
- Department of Medical Oncology, Ospedale Santa Maria Delle Croci, Ravenna AUSL Romagna, Italy
| | - Elena Fea
- Department of Medical Oncology, S. Croce E Carle Teaching Hospital, Cuneo, Italy
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliera, Universitaria Di Bologna, Bologna, Italy
| | - Ilario Giovanni Rapposelli
- Department of Medical Oncology, IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Massimiliano Salati
- Oncology Unit, University Hospital of Modena, Modena Cancer Centre, Modena, Italy
| | - Paolo Baili
- Department of Epidemiology and Data Science, Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rosalba Miceli
- Biostatistics for Clinical Research Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silva Ljevar
- Biostatistics for Clinical Research Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ilaria Cavallo
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisa Sottotetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian, 1, 20133, Milan, Italy
| | - Antonia Martinetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian, 1, 20133, Milan, Italy
| | - Michele Droz Dit Busset
- Department of Surgery, Division of HPB, General Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carlo Sposito
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Surgery, Division of HPB, General Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian, 1, 20133, Milan, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian, 1, 20133, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian, 1, 20133, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Vincenzo Mazzaferro
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Surgery, Division of HPB, General Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
230
|
Fischer RA, Ryan I, De La Torre K, Barnett C, Sehgal VS, Levy JB, Luke JJ, Poklepovic AS, Hurlbert MS. US physician perspective on the use of biomarker and ctDNA testing in patients with melanoma. Crit Rev Oncol Hematol 2024; 196:104289. [PMID: 38341119 DOI: 10.1016/j.critrevonc.2024.104289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024] Open
Abstract
New treatments have increased survival of patients with melanoma, and methods to monitor patients throughout the disease process are needed. Circulating tumor DNA (ctDNA) is a predictive and prognostic biomarker that may allow routine, real-time monitoring of disease status. We surveyed 44 US physicians to understand their preferences and practice patterns for biomarker and ctDNA testing in their patients with melanoma. Tumor biomarker testing was often ordered in stage IIIA-IV patients. Barriers to biomarker testing include insufficient tissue (60%) and lack of insurance coverage (54%). ctDNA testing was ordered by 16-18% of physicians for stages II-IV. Reasons for not using ctDNA testing included lack of prospective data (41%), ctDNA testing used for research only (18%), and others. Physicians (≥74%) believed that ctDNA assays could help with monitoring and treatment selection throughout the disease process. Physicians consider ctDNA testing potentially valuable for clinical decision-making but cited concerns that should be addressed.
Collapse
Affiliation(s)
- Rachel A Fischer
- Melanoma Research Alliance, 730 15th St NW, Washington, DC 20005, USA
| | - Isabel Ryan
- Melanoma Research Alliance, 730 15th St NW, Washington, DC 20005, USA
| | | | - Cody Barnett
- Melanoma Research Alliance, 730 15th St NW, Washington, DC 20005, USA
| | - Viren S Sehgal
- Melanoma Research Alliance, 730 15th St NW, Washington, DC 20005, USA
| | - Joan B Levy
- Melanoma Research Alliance, 730 15th St NW, Washington, DC 20005, USA
| | - Jason J Luke
- Cancer Immunotherapeutics Center, University of Pittsburgh Medical Center, 5150 Centre Avenue, Pittsburgh, PA 15232, USA
| | - Andrew S Poklepovic
- Virginia Commonwealth University Health System Massey Cancer Center, 401 College Street, Richmond, VA 23298-0037, USA
| | - Marc S Hurlbert
- Melanoma Research Alliance, 730 15th St NW, Washington, DC 20005, USA.
| |
Collapse
|
231
|
Amonkar MM, Abderhalden LA, Fox GE, Frederickson AM, Grira T, Gozman A, Malhotra U, Malbecq W, Akers KG. Clinical outcomes for previously treated patients with advanced biliary tract cancer: a meta-analysis. Future Oncol 2024; 20:863-876. [PMID: 38353044 DOI: 10.2217/fon-2023-0006] [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: 01/03/2023] [Accepted: 01/30/2024] [Indexed: 03/27/2024] Open
Abstract
Aim: A systematic review and meta-analysis were performed to evaluate the efficacy of treatments for previously treated advanced biliary tract cancer (BTC) patients. Materials & methods: Databases were searched for studies evaluating treatments for advanced (unresectable and/or metastatic) BTC patients who progressed on prior therapy. Pooled estimates of objective response rate (ORR), median overall survival (OS) and median progression-free survival (PFS) were calculated using random effects meta-analysis. Results: Across 31 studies evaluating chemotherapy or targeted treatment regimens in an unselected advanced BTC patient population, pooled ORR was 6.9%, median OS was 6.6 months and median PFS was 3.2 months. Conclusion: The efficacy of conventional treatments for previously treated advanced BTC patients is poor and could be improved by novel therapies.
Collapse
|
232
|
Pihlmann Kristensen M, Korsgaard U, Timm S, Hansen TF, Zlobec I, Hager H, Kjær-Frifeldt S. The prognostic value of tumor budding in a thoroughly characterized stage II colon cancer population in the context of a national screening program. Hum Pathol 2024; 146:15-22. [PMID: 38428823 DOI: 10.1016/j.humpath.2024.02.010] [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: 12/22/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024]
Abstract
Tumor budding as a prognostic marker in colorectal cancer has not previously been investigated in a cohort of screened stage II colon cancer patients. We assessed the prognostic significance of tumor budding in a thoroughly characterized stage II colon cancer population comprising surgically resected patients in the Region of Southern Denmark from 2014 to 2016. Tumors were re-staged according to the 8th edition of UICC TNM Classification, undergoing detailed histopathological evaluation and tumor budding assessment following guidelines from the International Tumor Budding Consensus Conference. Prognostic evaluation utilized Kaplan-Meier curves, log-rank tests, and Cox proportional hazard models for time to recurrence (TTR), recurrence-free survival (RFS), and overall survival (OS). Out of 497 patients, 20% were diagnosed through the national colorectal cancer screening program. High-grade tumor budding (Bd3) was found in 19% of tumors and was associated with glandular subtype, perineural invasion, mismatch repair proficient tumors, and tumor recurrence (p < 0.001, p < 0.001, p = 0.045, and p = 0.007 respectively). In multivariable Cox regression, high-grade budding was a significant prognostic factor for TTR compared to low-grade (Bd3 HR 2.617; p = 0.007). An association between tumor budding groups and RFS was observed, and the difference was significant in univariable analysis for high-grade compared to low-grade tumor budding (Bd3 HR 1.461; p = 0.041). No significant differences were observed between tumor budding groups and OS. High-grade tumor budding is a predictor of recurrence in a screened population of patients with stage II colon cancer and should be considered a high-risk factor in a shared decision-making process when stratifying patients to adjuvant chemotherapy.
Collapse
Affiliation(s)
- Maria Pihlmann Kristensen
- Department of Pathology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark; Colorectal Cancer Center South, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark.
| | - Ulrik Korsgaard
- Department of Pathology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark; Colorectal Cancer Center South, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Signe Timm
- Institute of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark; Colorectal Cancer Center South, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark; Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Torben Frøstrup Hansen
- Institute of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark; Colorectal Cancer Center South, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark; Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Inti Zlobec
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | - Henrik Hager
- Department of Pathology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark; Colorectal Cancer Center South, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark; Department of Pathology, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Sanne Kjær-Frifeldt
- Department of Pathology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark; Colorectal Cancer Center South, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| |
Collapse
|
233
|
Feng D, Wang J, Xiao Y, Wu R, Li D, Tuo Z, Yu Q, Ye L, MIYAMOTO A, Yoo KH, Wei W, Ye X, Zhang C, Han P. SKA3 targeted therapies in cancer precision surgery: bridging bench discoveries to clinical applications - review article. Int J Surg 2024; 110:2323-2337. [PMID: 38241327 PMCID: PMC11020031 DOI: 10.1097/js9.0000000000001123] [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: 09/11/2023] [Accepted: 01/09/2024] [Indexed: 01/21/2024]
Abstract
Spindle and kinetochore-associated complex subunit 3 (SKA3) is a microtubule-binding subcomplex of the outer kinetochore, which plays a vital role in proper chromosomal segregation and cell division. Recently, SKA3 have been demonstrated its oncogenic role of tumorigenesis and development in cancers. In this review, the authors comprehensively deciphered SKA3 in human cancer from various aspects, including bibliometrics, pan-cancer analysis, and narrative summary. The authors also provided the top 10 predicted drugs targeting SKA3. The authors proposed that SKA3 was a potential target and brought new therapeutic opportunities for cancer patients.
Collapse
Affiliation(s)
- Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| | - Yuhan Xiao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo City, Zhejiang Province
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
| | - Akira MIYAMOTO
- Department of Rehabilitation, West Kyushu University, Japan
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, South Korea
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| | - Xing Ye
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Chi Zhang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou
| | - Ping Han
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu
| |
Collapse
|
234
|
Hackenbruch C, Bauer J, Heitmann JS, Maringer Y, Nelde A, Denk M, Zieschang L, Kammer C, Federmann B, Jung S, Martus P, Malek NP, Nikolaou K, Salih HR, Bitzer M, Walz JS. FusionVAC22_01: a phase I clinical trial evaluating a DNAJB1-PRKACA fusion transcript-based peptide vaccine combined with immune checkpoint inhibition for fibrolamellar hepatocellular carcinoma and other tumor entities carrying the oncogenic driver fusion. Front Oncol 2024; 14:1367450. [PMID: 38606105 PMCID: PMC11007196 DOI: 10.3389/fonc.2024.1367450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/13/2024] [Indexed: 04/13/2024] Open
Abstract
The DNAJB1-PRKACA fusion transcript was identified as the oncogenic driver of tumor pathogenesis in fibrolamellar hepatocellular carcinoma (FL-HCC), also known as fibrolamellar carcinoma (FLC), as well as in other tumor entities, thus representing a broad target for novel treatment in multiple cancer entities. FL-HCC is a rare primary liver tumor with a 5-year survival rate of only 45%, which typically affects young patients with no underlying primary liver disease. Surgical resection is the only curative treatment option if no metastases are present at diagnosis. There is no standard of care for systemic therapy. Peptide-based vaccines represent a low side-effect approach relying on specific immune recognition of tumor-associated human leucocyte antigen (HLA) presented peptides. The induction (priming) of tumor-specific T-cell responses against neoepitopes derived from gene fusion transcripts by peptide-vaccination combined with expansion of the immune response and optimization of immune function within the tumor microenvironment achieved by immune-checkpoint-inhibition (ICI) has the potential to improve response rates and durability of responses in malignant diseases. The phase I clinical trial FusionVAC22_01 will enroll patients with FL-HCC or other cancer entities carrying the DNAJB1-PRKACA fusion transcript that are locally advanced or metastatic. Two doses of the DNAJB1-PRKACA fusion-based neoepitope vaccine Fusion-VAC-XS15 will be applied subcutaneously (s.c.) with a 4-week interval in combination with the anti-programmed cell death-ligand 1 (PD-L1) antibody atezolizumab starting at day 15 after the first vaccination. Anti-PD-L1 will be applied every 4 weeks until end of the 54-week treatment phase or until disease progression or other reason for study termination. Thereafter, patients will enter a 6 months follow-up period. The clinical trial reported here was approved by the Ethics Committee II of the University of Heidelberg (Medical faculty of Mannheim) and the Paul-Ehrlich-Institute (P-00540). Clinical trial results will be published in peer-reviewed journals. Trial registration numbers EU CT Number: 2022-502869-17-01 and ClinicalTrials.gov Registry (NCT05937295).
Collapse
Affiliation(s)
- Christopher Hackenbruch
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Jens Bauer
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Jonas S. Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Yacine Maringer
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Annika Nelde
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Monika Denk
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Lisa Zieschang
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Christine Kammer
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Birgit Federmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Susanne Jung
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Peter Martus
- Institute for Medical Biometrics and Clinical Epidemiology, University Hospital Tübingen, Tübingen, Germany
| | - Nisar P. Malek
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
- Center for Personalized Medicine, University of Tübingen, Tübingen, Germany
- The M3 Research Institute, University of Tübingen, Tübingen, Germany
| | - Konstantin Nikolaou
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Helmut R. Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Michael Bitzer
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
- Center for Personalized Medicine, University of Tübingen, Tübingen, Germany
| | - Juliane S. Walz
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| |
Collapse
|
235
|
Tchilikidi KY. Ex vivo liver resection and auto-transplantation and special systemic therapy in perihilar cholangiocarcinoma treatment. World J Gastrointest Surg 2024; 16:635-640. [PMID: 38577079 PMCID: PMC10989340 DOI: 10.4240/wjgs.v16.i3.635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/26/2023] [Accepted: 02/18/2024] [Indexed: 03/22/2024] Open
Abstract
This editorial contains comments on the article "Systematic sequential therapy for ex vivo liver resection and autotransplantation: A case report and review of literature" in the recent issue of World Journal of Gastrointestinal Surgery. It points out the actuality and importance of the article and focuses primarily on the role and place of ex vivo liver resection and autotransplantation (ELRAT) and systemic therapy, underlying molecular mechanisms for targeted therapy in perihilar cholangiocarcinoma (pCCA) management. pCCA is a tough malignancy with a high proportion of advanced disease at the time of diagnosis. The only curative option is radical surgery. Surgical excision and reconstruction become extremely complicated and not always could be performed even in localized disease. On the other hand, ELRAT takes its place among surgical options for carefully selected pCCA patients. In advanced disease, systemic therapy becomes a viable option to prolong survival. This editorial describes current possibilities in chemotherapy and reveals underlying mechanisms and projections in targeted therapy with kinase inhibitors and immunotherapy in both palliative and adjuvant settings. Fibroblast grow factor and fibroblast grow factor receptor, human epidermal growth factor receptor 2, isocitrate dehydrogenase, and protein kinase cAMP activated catalytic subunit alpha (PRKACA) and beta (PRKACB) pathways have been actively investigated in CCA in last years. Several agents were introduced and approved by the Food and Drug Administration. They all demonstrated meaningful activity in CCA patients with no global change in outcomes. That is why every successfully treated patient counts, especially those with advanced disease. In conclusion, pCCA is still hard to treat due to late diagnosis and extremely complicated surgical options. ELRAT also brings some hope, but it could be performed in very carefully selected patients. Advanced disease requires systemic anticancer treatment, which is supposed to be individualized according to the genetic and molecular features of cancer cells. Targeted therapy in combination with chemo-immunotherapy could be effective in susceptible patients.
Collapse
Affiliation(s)
- Konstantin Y Tchilikidi
- Department of Surgery with Postgraduate Education, Altai State Medical University, Barnaul 656031, Russia
| |
Collapse
|
236
|
Wang W, Dong L, Lv H, An Y, Zhang C, Zheng Z, Guo Y, He L, Wang L, Wang J, Shi X, Li N, Zheng M. Downregulating miRNA-199a-5p exacerbates fluorouracil-induced cardiotoxicity by activating the ATF6 signaling pathway. Aging (Albany NY) 2024; 16:5916-5928. [PMID: 38536006 PMCID: PMC11042954 DOI: 10.18632/aging.205679] [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: 10/25/2023] [Accepted: 02/27/2024] [Indexed: 04/23/2024]
Abstract
BACKGROUND Fluorouracil (5-FU) might produce serious cardiac toxic reactions. miRNA-199a-5p is a miRNA primarily expressed in myocardial cells and has a protective effect on vascular endothelium. Under hypoxia stress, the expression level of miRNA-199a-5p was significantly downregulated and is closely related to cardiovascular events such as coronary heart disease, heart failure, and hypertension. We explored whether 5-FU activates the endoplasmic reticulum stress ATF6 pathway by regulating the expression of miRNA-199a-5p in cardiac toxicity. METHODS This project established a model of primary cardiomyocytes derived from neonatal rats and treated them with 5-FU in vitro. The expression of miRNA-199a-5p and its regulation were explored in vitro and in vivo. RESULTS 5-FU decreases the expression of miRNA-199a-5p in cardiomyocytes, activates the endoplasmic reticulum stress ATF6 pathway, and increases the expression of GRP78 and ATF6, affecting the function of cardiomyocytes, and induces cardiac toxicity. The rescue assay further confirmed that miRNA-199a-5p supplementation can reduce the cardiotoxicity caused by 5-FU, and its protective effect on cardiomyocytes depends on the downregulation of the endoplasmic reticulum ATF6 signaling pathway. CONCLUSIONS 5-FU can down-regulate expression of miRNA-199a-5p, then activate the endoplasmic reticulum stress ATF6 pathway, increase the expression of GRP78 and ATF6, affect the function of cardiomyocytes, and induce cardiac toxicity.
Collapse
Affiliation(s)
- Wei Wang
- Department of Oncology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031 Hebei, China
| | - Liang Dong
- Department of Cardiology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031, Hebei, China
| | - Hengxu Lv
- Department of Oncology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031 Hebei, China
| | - Yonghui An
- Department of Oncology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031 Hebei, China
| | - Changwang Zhang
- Department of Oncology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031 Hebei, China
| | - Zheng Zheng
- Department of Oncology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031 Hebei, China
| | - Ying Guo
- Department of Oncology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031 Hebei, China
| | - Li He
- Department of Oncology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031 Hebei, China
| | - Libin Wang
- Department of Oncology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031 Hebei, China
| | - Jinmei Wang
- Department of Oncology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031 Hebei, China
| | - Xinlei Shi
- Department of Oncology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031 Hebei, China
| | - Na Li
- Department of Oncology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031 Hebei, China
| | - Mingqi Zheng
- Department of Cardiology, The First Hospital of Hebei Medical University, Yuhua, Shijiazhuang 050031, Hebei, China
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang 050031, Hebei, China
| |
Collapse
|
237
|
Li W, Cheng X, Zhu G, Hu Y, Wang Y, Niu Y, Li H, Aierken A, Li J, Feng L, Liu G. A review of chemotherapeutic drugs-induced arrhythmia and potential intervention with traditional Chinese medicines. Front Pharmacol 2024; 15:1340855. [PMID: 38572424 PMCID: PMC10987752 DOI: 10.3389/fphar.2024.1340855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Significant advances in chemotherapy drugs have reduced mortality in patients with malignant tumors. However, chemotherapy-related cardiotoxicity increases the morbidity and mortality of patients, and has become the second leading cause of death after tumor recurrence, which has received more and more attention in recent years. Arrhythmia is one of the common types of chemotherapy-induced cardiotoxicity, and has become a new risk related to chemotherapy treatment, which seriously affects the therapeutic outcome in patients. Traditional Chinese medicine has experienced thousands of years of clinical practice in China, and has accumulated a wealth of medical theories and treatment formulas, which has unique advantages in the prevention and treatment of malignant diseases. Traditional Chinese medicine may reduce the arrhythmic toxicity caused by chemotherapy without affecting the anti-cancer effect. This paper mainly discussed the types and pathogenesis of secondary chemotherapeutic drug-induced arrhythmia (CDIA), and summarized the studies on Chinese medicine compounds, Chinese medicine Combination Formula and Chinese medicine injection that may be beneficial in intervention with secondary CDIA including atrial fibrillation, ventricular arrhythmia and sinus bradycardia, in order to provide reference for clinical prevention and treatment of chemotherapy-induced arrhythmias.
Collapse
Affiliation(s)
- Weina Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaozhen Cheng
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guanghui Zhu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Hu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Yunhan Wang
- Henan Province Hospital of Traditional Chinese Medicine (The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, Henan, China
| | - Yueyue Niu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongping Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aikeremu Aierken
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ling Feng
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guifang Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
238
|
Nagy A, Börzsei D, Hoffmann A, Török S, Veszelka M, Almási N, Varga C, Szabó R. A Comprehensive Overview on Chemotherapy-Induced Cardiotoxicity: Insights into the Underlying Inflammatory and Oxidative Mechanisms. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07574-0. [PMID: 38492161 DOI: 10.1007/s10557-024-07574-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
While oncotherapy has made rapid progress in recent years, side effects of anti-cancer drugs and treatments have also come to the fore. These side effects include cardiotoxicity, which can cause irreversible cardiac damages with long-term morbidity and mortality. Despite the continuous in-depth research on anti-cancer drugs, an improved knowledge of the underlying mechanisms of cardiotoxicity are necessary for early detection and management of cardiac risk. Although most reviews focus on the cardiotoxic effect of a specific individual chemotherapeutic agent, the aim of our review is to provide comprehensive insight into various agents that induced cardiotoxicity and their underlying mechanisms. Characterization of these mechanisms are underpinned by research on animal models and clinical studies. In order to gain insight into these complex mechanisms, we emphasize the role of inflammatory processes and oxidative stress on chemotherapy-induced cardiac changes. A better understanding and identification of the interplay between chemotherapy and inflammatory/oxidative processes hold some promise to prevent or at least mitigate cardiotoxicity-associated morbidity and mortality among cancer survivors.
Collapse
Affiliation(s)
- András Nagy
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Denise Börzsei
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Alexandra Hoffmann
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Szilvia Török
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Médea Veszelka
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Nikoletta Almási
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Csaba Varga
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Renáta Szabó
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary.
| |
Collapse
|
239
|
Feng SH, Zhao B, Zhan X, Li RH, Yang Q, Wang SM, Li A. Quercetin-induced pyroptosis in colon cancer through NEK7-mediated NLRP3 inflammasome-GSDMD signaling pathway activation. Am J Cancer Res 2024; 14:934-958. [PMID: 38590424 PMCID: PMC10998754 DOI: 10.62347/mkan3550] [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: 12/29/2023] [Accepted: 02/27/2024] [Indexed: 04/10/2024] Open
Abstract
Pyroptosis, a gasdermin-mediated lytic cell death, is a new hotspot topic in cancer research, and induction of tumor pyroptosis has emerged as a new target in cancer management. Quercetin (Que), a natural substance, demonstrates promising anticancer action. However, further information is required to fully comprehend the function and mechanism of Que in pyroptosis in colon cancer. This study revealed the underlying mechanism of Que-induced pyroptosis in colon cancer in vitro and in vivo. Que inhibited colon cancer cell growth through gasdermin D (GSDMD)-mediated pyroptosis. Depletion of GSDMD, rather than gasdermin E (GSDME), reversed the cytotoxic effects of Que on colon cancer cells. Que treatment upregulated NIMA-related kinase 7 (NEK7) protein expression, thus facilitating the assembly of the NLRP3 inflammasome and cleavage of GSDMD. NEK7 silencing resulted in colon cancer cell growth in vitro and in vivo. Mechanistically, NEK7 depression restrained the activation of the NLRP3 inflammasome-GSDMD pathway, thus attenuating pyroptosis triggered by Que in colon cancer cells. Furthermore, lower NEK7 and NLRP3 expression levels indicated colon cancer progression. Our results unveiled a novel pattern of anti-colon cancer activity of Que, and activation of NEK7-mediated pyroptosis is potentially a promising therapeutic target for colon cancer, which provides novel experimental proof for the clinical application of Que.
Collapse
Affiliation(s)
- Shi-Han Feng
- Department of Traditional Chinese Medicine, Yong Chuan Hospital of Chongqing Medical University, Chongqing Medical UniversityChongqing 402160, P. R. China
| | - Bin Zhao
- Department of Traditional Chinese Medicine, Yong Chuan Hospital of Chongqing Medical University, Chongqing Medical UniversityChongqing 402160, P. R. China
| | - Xue Zhan
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical UniversityChongqing 400016, P. R. China
| | - Rong-Heng Li
- Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical UniversityChongqing 400042, P. R. China
| | - Qian Yang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical UniversityChongqing 400016, P. R. China
| | - Shu-Mei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical UniversityChongqing 400016, P. R. China
| | - Ao Li
- Department of Traditional Chinese Medicine, Yong Chuan Hospital of Chongqing Medical University, Chongqing Medical UniversityChongqing 402160, P. R. China
| |
Collapse
|
240
|
Ikezawa K, Urabe M, Kai Y, Takada R, Akita H, Nagata S, Ohkawa K. Comprehensive review of pancreatic acinar cell carcinoma: epidemiology, diagnosis, molecular features and treatment. Jpn J Clin Oncol 2024; 54:271-281. [PMID: 38109477 PMCID: PMC10925851 DOI: 10.1093/jjco/hyad176] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023] Open
Abstract
Pancreatic acinar cell carcinoma is a rare form (0.2-4.3%) of pancreatic neoplasm with unique clinical and molecular characteristics, which largely differ from pancreatic ductal adenocarcinoma. Pancreatic acinar cell carcinoma occurs more frequently in males and can occur in children. Serum lipase is elevated in 24-58% of patients with pancreatic acinar cell carcinoma. Pancreatic acinar cell carcinomas tend to be large at diagnosis (median tumour size: ~5 cm) and are frequently located in the pancreas head. Radiologically, pancreatic acinar cell carcinoma generally exhibits a solid appearance; however, necrosis, cystic changes and intratumoral haemorrhage can occur in larger lesions. Immunostaining is essential for the definitive diagnosis of pancreatic acinar cell carcinoma. Compared with pancreatic ductal adenocarcinoma, pancreatic acinar cell carcinoma has a more favourable prognosis. Although radical surgery is recommended for patients with pancreatic acinar cell carcinoma who do not have distant metastases, the recurrence rate is high. The effectiveness of adjuvant therapy for pancreatic acinar cell carcinoma is unclear. The response to FOLFIRINOX is generally favourable, and some patients achieve a complete response. Pancreatic acinar cell carcinoma has a different genomic profile compared with pancreatic ductal adenocarcinoma. Although genomic analyses have shown that pancreatic acinar cell carcinoma rarely has KRAS, TP53 and CDKN2A mutations, it has a higher prevalence of homologous recombination-related genes, including BRCA1/2 and ATM, than pancreatic ductal adenocarcinoma, suggesting high sensitivity to platinum-containing regimens and PARP inhibitors. Targeted therapies for genomic alternations are beneficial. Therefore, genetic testing is important for patients with pancreatic acinar cell carcinoma to choose the optimal therapeutic strategy.
Collapse
Affiliation(s)
- Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Makiko Urabe
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Yugo Kai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Ryoji Takada
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Hirofumi Akita
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Shigenori Nagata
- Department of Diagnostic Pathology and Cytology, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
241
|
Shi H, Duan L, Tong L, Pu P, Wei L, Wang L, Hu D, Tang H. Research Progress on Flavonoids in Traditional Chinese Medicine to Counteract Cardiotoxicity Associated with Anti-Tumor Drugs. Rev Cardiovasc Med 2024; 25:74. [PMID: 39076949 PMCID: PMC11263839 DOI: 10.31083/j.rcm2503074] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/12/2023] [Accepted: 11/21/2023] [Indexed: 07/31/2024] Open
Abstract
The development of anti-tumor drugs has notably enhanced the survival rates and quality of life for patients with malignant tumors. However, the side effects of these drugs, especially cardiotoxicity, significantly limit their clinical application. The cardiotoxicity associated with anti-tumor drugs has been a subject of extensive attention and research. Traditional to mitigate these side effects have included reducing drug dosages, shortening treatment duration, modifying administration methods, and opting for drugs with lower toxicity. However, either approach may potentially compromise the anti-tumor efficacy of the medications. Therefore, exploring other effective methods for anti-cardiotoxicity will be the focus of future research. The potential of traditional Chinese medicine (TCM) in managing cardiovascular diseases and cancer treatment has gained widespread recognition. TCM is valued for its minimal side effects, affordability, and accessibility, offering promising avenues in the prevention and treatment of cardiotoxicity caused by anti-tumor drugs. Among its constituents, flavonoids, which are present in many TCMs, are particularly notable. These monomeric compounds with distinct structural components have been shown to possess both cardiovascular protective properties and anti-tumor capabilities. In this discussion, we will delve into the classification of anti-tumor drugs and explore the underlying mechanisms of their associated cardiotoxicity. Additionally, we will examine flavonoids found in TCM and investigate their mechanisms of cardiovascular protection. This will include an analysis of how these natural compounds can mitigate the cardiac side effects of anti-tumor therapies while potentially enhancing overall patient health and treatment outcomes.
Collapse
Affiliation(s)
- Hongwei Shi
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
- Department of Oncology, Renmin Hospital of Wuhan University, 430064 Wuhan, Hubei, China
| | - Lian Duan
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Li Tong
- Department of Pharmacy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Peng Pu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Lai Wei
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Linlin Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Desheng Hu
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Heng Tang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| |
Collapse
|
242
|
Elshami M, Ammori JB, Hardacre JM, Selfridge JE, Bajor D, Mohamed A, Chakrabarti S, Mahipal A, Winter JM, Ocuin LM. Surgical Resection Alone is Associated With Higher Long-Term Survival Than Multiagent Chemotherapy Alone for Patients With Localized Biliary Tract Cancers. J Surg Res 2024; 295:705-716. [PMID: 38141457 DOI: 10.1016/j.jss.2023.11.067] [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: 03/29/2023] [Revised: 07/17/2023] [Accepted: 11/12/2023] [Indexed: 12/25/2023]
Abstract
INTRODUCTION We compared long-term survival of patients with localized biliary tract cancers (BTCs) treated with either surgical resection or multiagent chemotherapy. METHODS Patients with localized BTC [gallbladder adenocarcinoma, extrahepatic cholangiocarcinoma, intrahepatic cholangiocarcinoma] were identified within the National Cancer Database (2010-2017). Piecewise-constant hazard modeling was used to estimate hazard ratios (HRs) at prespecified intervals: 0-30 d, 31-60 d, 61-90 d, and >90 d post-treatment. RESULTS A total of 5988 patients with localized BTC were identified: 2697 (45.0%) received multiagent chemotherapy and 3291 (55.0%) underwent surgical resection. Patients with gallbladder adenocarcinoma or extrahepatic cholangiocarcinoma who were treated with surgical resection had an associated decline in overall survival (OS) as compared to those treated with multiagent chemotherapy within 0-30 d of treatment initiation (gallbladder adenocarcinoma [adjusted HR = 3.94, 95% confidence interval [CI]: 1.77-8.80]; extrahepatic cholangiocarcinoma [adjusted HR = 4.88, 95% CI: 2.76-8.61]). However, there was an associated improvement in OS for patients treated with surgical resection after 90 d from treatment initiation (gallbladder adenocarcinoma [adjusted HR = 0.36, 95% CI: 0.28-0.46]; extrahepatic cholangiocarcinoma [adjusted HR = 0.27, 95% CI: 0.24-0.32]). Among patients with intrahepatic cholangiocarcinoma, those who underwent surgical resection had an associated improvement in OS at 31-60 d (adjusted HR = 0.63, 95% CI: 0.40-0.99) and a further associated increase in OS at 61-90 d (adjusted HR = 0.34, 95% CI: 0.21-0.54) and after 90 d (HR = 0.23, 95% CI: 0.21-0.27) of treatment initiation. CONCLUSIONS For patients with localized BTC, surgical resection alone is associated with improved long-term survival outcomes compared to multiagent chemotherapy alone.
Collapse
Affiliation(s)
- Mohamedraed Elshami
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - John B Ammori
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Jeffrey M Hardacre
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - J Eva Selfridge
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - David Bajor
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Amr Mohamed
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Sakti Chakrabarti
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Amit Mahipal
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Jordan M Winter
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Lee M Ocuin
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, Ohio.
| |
Collapse
|
243
|
Popovich SM, Vetter TR. Preoperative Management of the Adult Oncology Patient. Anesthesiol Clin 2024; 42:145-158. [PMID: 38278586 DOI: 10.1016/j.anclin.2023.07.004] [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: 01/28/2024]
Abstract
Anesthesiologists are experiencing first-hand the aging population, given older patients more frequently presenting for surgery, often with geriatric syndromes influencing their anesthetic management. The overall incidence and health burden of cancer morbidity and mortality are also rapidly increasing worldwide. This growth in the cancer population, along with the associated risk factors and comorbidities often accompanying a cancer diagnosis, underscores the need for anesthesiologists to become well versed in the preoperative evaluation and management of the adult patient with cancer. This article will focus on the unique challenges and opportunities for the anesthesiologist caring for the adult oncology patient presenting for surgery.
Collapse
Affiliation(s)
- Shannon M Popovich
- Department of Anesthesiology and Perioperative Medicine, Division of Anesthesiology, Critical Care Medicine and Pain Medicine, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Thomas R Vetter
- Department of Surgery and Perioperative Care, Dell Medical School at The University of Texas at Austin, Health Discovery Building, Room 6.812, 1701 Trinity Street, Austin, TX 78712-1875, USA.
| |
Collapse
|
244
|
Elliott JA, Guinan E, Reynolds JV. Measurement and optimization of perioperative risk among patients undergoing surgery for esophageal cancer. Dis Esophagus 2024; 37:doad062. [PMID: 37899136 PMCID: PMC10906714 DOI: 10.1093/dote/doad062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/28/2023] [Indexed: 10/31/2023]
Abstract
Esophagectomy is an exemplar of complex oncological surgery and is associated with a relatively high risk of major morbidity and mortality. In the modern era, where specific complications are targeted in prevention and treatment pathways, and where the principles of enhanced recovery after surgery are espoused, optimum outcomes are targeted via a number of approaches. These include comprehensive clinical and physiological risk assessment, specialist perioperative care by a high-volume team, and multimodal inputs throughout the patient journey that aim to preserve or restore nutritional deficits, muscle mass and function.
Collapse
Affiliation(s)
- Jessie A Elliott
- Trinity St. James’s Cancer Institute, Trinity College Dublin and St. James’s Hospital, Dublin, Ireland
| | - Emer Guinan
- Trinity St. James’s Cancer Institute, Trinity College Dublin and St. James’s Hospital, Dublin, Ireland
| | - John V Reynolds
- Trinity St. James’s Cancer Institute, Trinity College Dublin and St. James’s Hospital, Dublin, Ireland
| |
Collapse
|
245
|
Kabat M, Padalkar R, Hazaveh S, Joseph V, Feigenblum D, Sadikot S. Capecitabine-induced-coronary-vasospasm leading to polymorphic ventricular tachycardia and cardiac arrest. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:11. [PMID: 38414072 PMCID: PMC10898064 DOI: 10.1186/s40959-024-00214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/21/2024] [Indexed: 02/29/2024]
Abstract
Capecitabine, a pro-drug of 5-fluorouracil, is commonly used in the treatment of breast and colorectal cancer. Its side effects, including nausea, vomiting, diarrhea, fatigue, loss of appetite, and bone marrow suppression, are well recognized. However, coronary vasospasm represents a less commonly recognized but significant complication of fluoropyrimidine-based therapies such as capecitabine. Proposed mechanisms for this adverse effect complication include direct endothelium-independent vasoconstriction, activation of protein kinase C, and activation of the cyclooxygenase pathway. In this report, we present a case of capecitabine-induced coronary vasospasm leading to progressive, focal ST-elevations, myocardial ischemia, and subsequently polymorphic ventricular tachycardia. These events were captured on telemetry, in a male in his early 40s, diagnosed with stage IIIB sigmoid colon cancer. Notably, the patient had no pre-existing coronary artery disease or other cardiovascular risk factors. Upon diagnosis, the patient was initiated on a calcium channel blocker, verapamil, to mitigate further coronary vasospasm events. After thorough discussions that prioritized the patient's input and values, an implantable cardioverter-defibrillator was placed subcutaneously. Following discharge, the patient restarted capecitabine therapy along with verapamil prophylaxis and did not experience any subsequent shocks from his ICD as assessed during his outpatient follow-up visits. This case emphasizes the need to involve patients in decision-making processes, especially when managing unexpected and serious complications, to ensure treatments align with their quality of life and personal preferences.
Collapse
Affiliation(s)
- Maciej Kabat
- Department of Internal Medicine, Hackensack University Medical Center, 30 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Roma Padalkar
- Department of Internal Medicine, Hackensack University Medical Center, 30 Prospect Avenue, Hackensack, NJ, 07601, USA
| | - Sara Hazaveh
- Department of Internal Medicine, Hackensack University Medical Center, 30 Prospect Avenue, Hackensack, NJ, 07601, USA
| | - Vladimir Joseph
- Department of Cardiology, Hackensack University Medical Center, 30 Prospect Avenue, Hackensack, NJ, 07601, USA
| | - David Feigenblum
- Department of Cardiac Electrophysiology, Hackensack University Medical Center, 30 Prospect Avenue, Hackensack, NJ, 07601, USA
| | - Sean Sadikot
- Department of Critical Care, Hackensack University Medical Center, 30 Prospect Avenue, Hackensack, NJ, 07601, USA
| |
Collapse
|
246
|
Cheng XF, Zhao F, Chen D, Liu FL. Current landscape of preoperative neoadjuvant therapies for initial resectable colorectal cancer liver metastasis. World J Gastroenterol 2024; 30:663-672. [PMID: 38515943 PMCID: PMC10950626 DOI: 10.3748/wjg.v30.i7.663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 12/22/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2024] Open
Abstract
Colorectal cancer liver metastasis (CRLM) presents a clinical challenge, and optimizing treatment strategies is crucial for improving patient outcomes. Surgical resection, a key element in achieving prolonged survival, is often linked to a heightened risk of recurrence. Acknowledging the potential benefits of preoperative neoadjuvant chemotherapy in managing resectable liver metastases, this approach has gained attention for its role in tumor downsizing, assessing biological behavior, and reducing the risk of postoperative recurrence. However, the use of neoadjuvant chemotherapy in initially resectable CRLM sparks ongoing debates. The balance between tumor reduction and the risk of hepatic injury, coupled with concerns about delaying surgery, necessitates a nuanced approach. This article explores recent research insights and draws upon the practical experiences at our center to address critical issues regarding considerations for initially resectable cases. Examining the criteria for patient selection and the judicious choice of neoadjuvant regimens are pivotal areas of discussion. Striking the right balance between maximizing treatment efficacy and minimizing adverse effects is imperative. The dynamic landscape of precision medicine is also reflected in the evolving role of gene testing, such as RAS/BRAF and PIK3CA, in tailoring neoadjuvant regimens. Furthermore, the review emphasizes the need for a multidisciplinary approach to navigate the complexities of CRLM. Integrating technical expertise and biological insights is crucial in refining neoadjuvant strategies. The management of progression following neoadjuvant chemotherapy requires a tailored approach, acknowledging the diverse biological behaviors that may emerge. In conclusion, this review aims to provide a comprehensive perspective on the considerations, challenges, and advancements in the use of neoadjuvant chemotherapy for initially resectable CRLM. By combining evidence-based insights with practical experiences, we aspire to contribute to the ongoing discourse on refining treatment paradigms for improved outcomes in patients with CRLM.
Collapse
Affiliation(s)
- Xiao-Fei Cheng
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Feng Zhao
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Dong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Fan-Long Liu
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
247
|
Stelzner S, Mehdorn M, Puffer E, Bleyl D, Kittner T, Rhode P, Gockel I, Mees ST. Sidedness is not a prognostic factor in an unselected cohort of patients with colon cancer but prognosis for caecal carcinoma is worse - A multivariate analysis of a large single institution database. Int J Colorectal Dis 2024; 39:27. [PMID: 38349566 PMCID: PMC10864445 DOI: 10.1007/s00384-023-04590-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 02/15/2024]
Abstract
PURPOSE Sidedness has emerged as a prognostic factor for metastatic colorectal cancer treated with modern systemic therapies. This study investigates whether it is also relevant for an unselected patient cohort including all stages. METHODS All consecutive patients admitted with colon cancer between 1995 and 2018 were retrieved from an institution-held database. Patients were divided into two cohorts. The first cohort included patients without distant metastases who were able to undergo curative resection. The second cohort presented with distant metastases (stage IV). Potentially prognostic factors were subjected to multivariate Cox Regression analysis. RESULTS Overall, 1,606 patients met the inclusion and exclusion criteria. An R0-resection was achieved in 1,222 patients without distant metastases. Five-year cause-specific survival rate was 89.3% for this group. There was no difference between right- and left-sided cancers (88.2% vs. 90.1%, p = 0.220). However, prognosis of caecal carcinoma was significantly worse than that of all other sites combined (83.5% vs. 90.2%, p = 0.007). In multivariate analysis, pT-category, pN-category, grading, vascular invasion, emergency operation, adjuvant chemotherapy, and caecal carcinoma remained as independent prognostic factors. In the 384 patients with stage IV-disease, 3-year overall survival for right- vs. left-sided cancers differed only in univariate analysis (17.7% vs. 28.6%, p = 0.013). CONCLUSION In non-metastatic colon cancer, location in the caecum is an independent prognostic factor. In unselected patients with stage IV colon cancer, sidedness was not found to be a prognostic factor. Differentiation into right- and left-sided tumors may be simplistic, and further studies on the biological behavior of different colonic sites are warranted.
Collapse
Affiliation(s)
- Sigmar Stelzner
- Department of General and Visceral Surgery, Dresden-Friedrichstadt General Hospital, Teaching Hospital of the Technical University Dresden, Friedrichstr. 41, D-01067, Dresden, Germany.
- Department of Visceral, Transplant, Thoracic, and Vascular Surgery, University Hospital of Leipzig, Liebigstr. 20, D-04103, Leipzig, Germany.
| | - Matthias Mehdorn
- Department of Visceral, Transplant, Thoracic, and Vascular Surgery, University Hospital of Leipzig, Liebigstr. 20, D-04103, Leipzig, Germany
| | - Erik Puffer
- Department of Pathology, Dresden-Friedrichstadt General Hospital, Teaching Hospital of the Technical University Dresden, Friedrichstr. 41, D-01067, Dresden, Germany
| | - Dorothea Bleyl
- Department of Medical Oncology, Dresden-Friedrichstadt General Hospital, Teaching Hospital of the Technical University Dresden, Friedrichstr. 41, D-01067, Dresden, Germany
| | - Thomas Kittner
- Department of Radiology, Dresden-Friedrichstadt General Hospital, Teaching Hospital of the Technical University Dresden, Friedrichstr. 41, D-01067, Dresden, Germany
| | - Philipp Rhode
- Department of Visceral, Transplant, Thoracic, and Vascular Surgery, University Hospital of Leipzig, Liebigstr. 20, D-04103, Leipzig, Germany
| | - Ines Gockel
- Department of Visceral, Transplant, Thoracic, and Vascular Surgery, University Hospital of Leipzig, Liebigstr. 20, D-04103, Leipzig, Germany
| | - Soeren T Mees
- Department of General and Visceral Surgery, Dresden-Friedrichstadt General Hospital, Teaching Hospital of the Technical University Dresden, Friedrichstr. 41, D-01067, Dresden, Germany
| |
Collapse
|
248
|
Su BC, Chen GY, Yang CM, Chuang WT, Lin MC, Hsu PL, Lee CW, Cheng CC, Wu SY, Pan BS, Yu HH. Impacts of hyperthermic chemotherapeutic agent on cytotoxicity, chemoresistance-related proteins and PD-L1 expression in human gastric cancer cells. Int J Hyperthermia 2024; 41:2310017. [PMID: 38350654 DOI: 10.1080/02656736.2024.2310017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/15/2024] Open
Abstract
Objective: Gastric cancer with peritoneal metastasis is considered to be final stage gastric cancer. One current treatment approach for this condition is combined cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (HIPEC). However, the therapeutic mechanisms of HIPEC remain largely undescribed. Method: In order to assess the cellular effects of HIPEC in vitro, we treated AGS human gastric adenocarcinoma cells with or without 5-fluorouracil (5-Fu) at 37 °C or at 43 °C (hyperthermic temperature) for 1 h followed by incubation at 37 °C for 23 h. The impacts of hyperthermia/5-Fu on apoptosis, cell survival signals, oxidative stress, chemoresistance-related proteins and programmed death-ligand 1 (PD-L1) expression were measured. Results: Our results showed that hyperthermia potentiates 5-Fu-mediated cytotoxicity in AGS cells. Furthermore, the combination of 5-Fu and hyperthermia reduces levels of both phosphorylated STAT3 and STAT3, while increasing the levels of phosphorylated Akt and ERK. In addition, 5-Fu/hyperthermia enhances reactive oxygen species and suppresses superoxide dismutase 1. Chemoresistance-related proteins, such as multidrug resistance 1 and thymidylate synthase, are also suppressed by 5-Fu/hyperthermia. Interestingly, hyperthermia enhances 5-Fu-mediated induction of glycosylated PD-L1, but 5-Fu-mediated upregulation of PD-L1 surface expression is prevented by hyperthermia. Conclusion: Taken together, our findings provide insights that may aid in the development of novel therapeutic strategies and enhanced therapeutic efficacy of HIPEC.
Collapse
Affiliation(s)
- Bor-Chyuan Su
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Guan-Yu Chen
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chun-Ming Yang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Wei-Ting Chuang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Meng-Chieh Lin
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Pei-Ling Hsu
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chu-Wan Lee
- Department of Nursing, National Tainan Junior College of Nursing, Tainan, Taiwan
| | - Chih-Cheng Cheng
- Division of General Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shih-Ying Wu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC, USA
| | - Bo-Syong Pan
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Hsin-Hsien Yu
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
249
|
Mesitskaya DF, Fashafsha ZZ, Poltavskaya MG, Andreev DA, Levshina AR, Sultygova EA, Gognieva D, Chomakhidze P, Kuznetsova N, Suvorov A, Marina I. S, Poddubskaya E, Novikova A, Bykova A, Kopylov P. A single-lead ECG based cardiotoxicity detection in patients on polychemotherapy. IJC HEART & VASCULATURE 2024; 50:101336. [PMID: 38304727 PMCID: PMC10831811 DOI: 10.1016/j.ijcha.2024.101336] [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: 09/26/2023] [Revised: 12/04/2023] [Accepted: 01/04/2024] [Indexed: 02/03/2024]
Abstract
Background Anti-cancer treatment can be fraught with cardiovascular complications, which is the most common cause of death among oncological survivors. Without appropriate cardiomonitoring during anti-cancer treatment, it becomes challenging to detect early signs of cardiovascular complications. In order to achieve higher survival rates, it is necessary to monitor oncological patients outpatiently after anti-cancer treatment administration. In this regard, we aim to evaluate the efficacy of single-lead ECG remote monitoring to detect cardiotoxicity in cancer patients with minimal cardiovascular diseases after the first cycle of polychemotherapy. Materials and methods The study included patients 162 patients over 18 years old with first diagnosed different types of solid tumors, planed for adjuvant (within 8 weeks after surgery) or neoadjuvant polychemotherapy. All patients were monitored, outpatiently, during 14-21 days (depending on the regimen of polychemotherapy) after polychemotherapy administration using single-lead ECG. Results QTc > 500 mc prolongation was detected in 8 patients (6.6 %), first-diagnosed arial fibrillation was detected in 11 patients (9 %) in period after chemotherapy administration. Moreover, left ventricular diastolic dysfunction using single-lead ECG after polychemotherapy was detected in 49 (40.1 %) patients with sensitivity 80 %, specificity 95 %, AUC 0.88 (95 % CI, 0.82-0.93). Conclusions The side effects of cancer treatment may cause life-threatening risks. Early identification of cardiotoxicity plays a vital role in the solution of this problem. Using portable devices to detect early cardiotoxicity is a simple, convenient and affordable screening method, that can be used for promptly observation of patients.
Collapse
Affiliation(s)
- Dinara F. Mesitskaya
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Zaki Z.A. Fashafsha
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maria G. Poltavskaya
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Denis A. Andreev
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Anna R. Levshina
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Elizaveta A. Sultygova
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Daria Gognieva
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Petr Chomakhidze
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Natalia Kuznetsova
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexander Suvorov
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sekacheva Marina I.
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
- Institute for Personalized Oncology, Center "Digital Biodesign and Personalized Healthcare" I.M. Sechenov First Moscow State Medical University Moscow, Russia Moscow, Russia
| | - Elena Poddubskaya
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Alena Novikova
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Aleksandra Bykova
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Philipp Kopylov
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
250
|
Henriksen TV, Demuth C, Frydendahl A, Nors J, Nesic M, Rasmussen MH, Reinert T, Larsen OH, Jaensch C, Løve US, Andersen PV, Kolbro T, Thorlacius-Ussing O, Monti A, Gögenur M, Kildsig J, Bondeven P, Schlesinger NH, Iversen LH, Gotschalck KA, Andersen CL. Unraveling the potential clinical utility of circulating tumor DNA detection in colorectal cancer-evaluation in a nationwide Danish cohort. Ann Oncol 2024; 35:229-239. [PMID: 37992872 DOI: 10.1016/j.annonc.2023.11.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/29/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Increasingly, circulating tumor DNA (ctDNA) is proposed as a tool for minimal residual disease (MRD) assessment. Digital PCR (dPCR) offers low analysis costs and turnaround times of less than a day, making it ripe for clinical implementation. Here, we used tumor-informed dPCR for ctDNA detection in a large colorectal cancer (CRC) cohort to evaluate the potential for post-operative risk assessment and serial monitoring, and how the metastatic site may impact ctDNA detection. Additionally, we assessed how altering the ctDNA-calling algorithm could customize performance for different clinical settings. PATIENTS AND METHODS Stage II-III CRC patients (N = 851) treated with a curative intent were recruited. Based on whole-exome sequencing on matched tumor and germline DNA, a mutational target was selected for dPCR analysis. Plasma samples (8 ml) were collected within 60 days after operation and-for a patient subset (n = 246)-every 3-4 months for up to 36 months. Single-target dPCR was used for ctDNA detection. RESULTS Both post-operative and serial ctDNA detection were prognostic of recurrence [hazard ratio (HR) = 11.3, 95% confidence interval (CI) 7.8-16.4, P < 0.001; HR = 30.7, 95% CI 20.2-46.7, P < 0.001], with a cumulative ctDNA detection rate of 87% at the end of sample collection in recurrence patients. The ctDNA growth rate was prognostic of survival (HR = 2.6, 95% CI 1.5-4.4, P = 0.001). In recurrence patients, post-operative ctDNA detection was challenging for lung metastases (4/21 detected) and peritoneal metastases (2/10 detected). By modifying the cut-off for calling a sample ctDNA positive, we were able to adjust the sensitivity and specificity of our test for different clinical contexts. CONCLUSIONS The presented results from 851 stage II-III CRC patients demonstrate that our personalized dPCR approach effectively detects MRD after operation and shows promise for serial ctDNA detection for recurrence surveillance. The ability to adjust sensitivity and specificity shows exciting potential to customize the ctDNA caller for specific clinical settings.
Collapse
Affiliation(s)
- T V Henriksen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus; Department of Clinical Medicine, Aarhus University, Aarhus
| | - C Demuth
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus; Department of Clinical Medicine, Aarhus University, Aarhus
| | - A Frydendahl
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus; Department of Clinical Medicine, Aarhus University, Aarhus
| | - J Nors
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus; Department of Clinical Medicine, Aarhus University, Aarhus
| | - M Nesic
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus; Department of Clinical Medicine, Aarhus University, Aarhus
| | - M H Rasmussen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus; Department of Clinical Medicine, Aarhus University, Aarhus
| | - T Reinert
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus; Department of Clinical Medicine, Aarhus University, Aarhus
| | - O H Larsen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus; Department of Clinical Medicine, Aarhus University, Aarhus
| | - C Jaensch
- Department of Surgery, Regional Hospital Gødstrup, Herning
| | - U S Løve
- Department of Surgery, Regional Hospital Viborg, Viborg
| | - P V Andersen
- Department of Surgery, Odense University Hospital, Odense
| | - T Kolbro
- Department of Surgery, Odense University Hospital, Svendborg
| | | | - A Monti
- Department of Surgery, North Denmark Regional Hospital Hjørring, Hjørring
| | - M Gögenur
- Center for Surgical Sciences, Zealand University Hospital, Køge
| | - J Kildsig
- Department of Surgery, Copenhagen University Hospital, Herlev
| | - P Bondeven
- Department of Surgery, Regional Hospital Randers, Randers
| | - N H Schlesinger
- Department of Surgery, Copenhagen University Hospital, Bispebjerg
| | - L H Iversen
- Department of Surgery, Aarhus University Hospital, Aarhus
| | - K A Gotschalck
- Department of Surgery, Regional Hospital Horsens, Horsens, Denmark
| | - C L Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus; Department of Clinical Medicine, Aarhus University, Aarhus.
| |
Collapse
|