1
|
Hu H, Zhang J, Li Y, Wang X, Wang Z, Wang H, Kang L, Liu P, Lan P, Wu X, Zhen Y, Pei H, Huang Z, Zhang H, Chen W, Zeng Y, Lai J, Wei H, Huang X, Chen J, Chen J, Tao K, Xu Q, Peng X, Liang J, Cai G, Ding K, Ding Z, Hu M, Zhang W, Tang B, Hong C, Cao J, Huang Z, Cao W, Li F, Wang X, Wang C, Huang Y, Zhao Y, Cai Y, Ling J, Xie X, Wu Z, Shi L, Ling L, Liu H, Wang J, Huang M, Deng Y. Neoadjuvant Chemotherapy With Oxaliplatin and Fluoropyrimidine Versus Upfront Surgery for Locally Advanced Colon Cancer: The Randomized, Phase III OPTICAL Trial. J Clin Oncol 2024; 42:2978-2988. [PMID: 38564700 DOI: 10.1200/jco.23.01889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/29/2023] [Accepted: 02/02/2024] [Indexed: 04/04/2024] Open
Abstract
PURPOSE The role of neoadjuvant chemotherapy (NAC) in colon cancer remains unclear. This trial investigated whether 3 months of modified infusional fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) or capecitabine and oxaliplatin (CAPOX) as NAC could improve outcomes in patients with locally advanced colon cancer versus upfront surgery. PATIENTS AND METHODS OPTICAL was a randomized, phase III trial in patients with clinically staged locally advanced colon cancer (T3 with extramural spread into the mesocolic fat ≥5 mm or T4). Patients were randomly assigned 1:1 to receive six preoperative cycles of mFOLFOX6 or four cycles of CAPOX, followed by surgery and adjuvant chemotherapy (NAC group), or immediate surgery and the physician's choice of adjuvant chemotherapy (upfront surgery group). The primary end point was 3-year disease-free survival (DFS) assessed in the modified intention-to-treat (mITT) population. RESULTS Between January 2016 and April 2021, of the 752 patients enrolled, 744 patients were included in the mITT analysis (371 in the NAC group; 373 in the upfront surgery group). At a median follow-up of 48.0 months (IQR, 46.0-50.1), 3-year DFS rates were 82.1% in the NAC group and 77.5% in the upfront surgery group (stratified hazard ratio [HR], 0.74 [95% CI, 0.54 to 1.03]). The R0 resection was achieved in 98% of patients who underwent surgery in both groups. Compared with upfront surgery, NAC resulted in a 7% pathologic complete response rate (pCR), significantly lower rates of advanced tumor staging (pT3-4: 77% v 94%), lymph node metastasis (pN1-2: 31% v 46%), and potentially improved overall survival (stratified HR, 0.44 [95% CI, 0.25 to 0.77]). CONCLUSION NAC with mFOLFOX6 or CAPOX did not show a significant DFS benefit. However, this neoadjuvant approach was safe, resulted in substantial pathologic downstaging, and appears to be a viable therapeutic option for locally advanced colon cancer.
Collapse
Affiliation(s)
- Huabin Hu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jianwei Zhang
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yunfeng Li
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Xiaozhong Wang
- Department of Gastrointestinal Surgery, Shantou Central Hospital, Shantou, People's Republic of China
| | - Ziqiang Wang
- Department of General Surgery, Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Liang Kang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ping Liu
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Ping Lan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaojian Wu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yunhuan Zhen
- Department of Colorectal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| | - Haiping Pei
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhongcheng Huang
- Department of General Surgery, Hunan Provincial People's Hospital, Changsha, People's Republic of China
| | - Hao Zhang
- Department of General Surgery, Dongguan Kanghua Hospital, Dongguan, People's Republic of China
| | - Wenbin Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yongming Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, People's Republic of China
| | - Jiajun Lai
- Department of Gastrointestinal Surgery, Yuebei People's Hospital, Shaoguan, People's Republic of China
| | - Hongbo Wei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xuefeng Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jiansi Chen
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Jigui Chen
- Department of Surgery, The Eighth Hospital of Wuhan, Wuhan, People's Republic of China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qingwen Xu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Xiang Peng
- Department of Gastrointestinal Surgery, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Junlin Liang
- Department of Coloproctological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Guanfu Cai
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangzhou, People's Republic of China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, Cancer Center, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Zhijie Ding
- Department of Gastrointestinal Surgery, Zhongshan Hospital, Xiamen University, Xiamen, People's Republic of China
| | - Ming Hu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Wei Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Bo Tang
- Department of General Surgery, The First Hospital Affiliated to Army Medical University, Chongqing, People's Republic of China
| | - Chuyuan Hong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jie Cao
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou, People's Republic of China
| | - Zonghai Huang
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Wuteng Cao
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Fangqian Li
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xinhua Wang
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Chao Wang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yandong Zhao
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yue Cai
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiayu Ling
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaoyu Xie
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zehua Wu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Lishuo Shi
- Clinical Research Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Ling
- Department of Medical Statistics, School of Public Health, and Center for Migrant Health Policy, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hao Liu
- Department of Biostatistics and Epidemiology, Biostatistics Shared Resource, Rutgers Cancer Institute of New Jersey, Rutgers School of Public Health, Brunswick, NJ
| | - Jianping Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Meijin Huang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
2
|
Suzumura H, Terauchi T, Yukisawa S, Kimata M, Shinozaki H. Curative Surgery After Neoadjuvant Chemotherapy for Locally Advanced Sigmoid Colon Cancer With Extensive Abdominal Wall Invasion: A Case Report. Cureus 2024; 16:e67444. [PMID: 39310573 PMCID: PMC11415604 DOI: 10.7759/cureus.67444] [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: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Locally advanced colon cancer (LACC) can be cured under an appropriate treatment strategy, but the decision on the treatment strategy is also important in terms of long-term prognosis. In cases with extensive abdominal wall involvement, it is especially important to secure adequate margins and repair abdominal wall defects. Recently, neoadjuvant chemotherapy (NAC) for LACC has shown promise in improving the chance of cure with tumor shrinkage. Herein, we report a case of curative surgery after NAC for locally advanced sigmoid colon cancer with extensive abdominal wall invasion. A 50-year-old woman visited our hospital with anemia and an abdominal mass. The diagnosis was LACC of the sigmoid colon with abdominal wall invasion (maximum size, 12 cm), and the clinical stage was stage IIIc (T4b[skin]N1bM0). Resection of the involved skin was expected to cause an extensive abdominal wall defect. At first, a colostomy was performed, followed by NAC with leucovorin, 5-fluorouracil, and oxaliplatin (FOLFOX). Ten cycles of chemotherapy were completed without severe adverse events, and the tumor shrank in size by approximately 39%. We performed a curative sigmoidectomy combined with abdominal wall resection with adequate margins. We reconstructed the abdominal wall defect using a left anterolateral thigh skin flap. Pathological examination revealed mucinous carcinoma involving the transverse colon and abdominal wall, with luminal narrowing in the sigmoid colon. The surgical margins were negative, and the tumor was considered to have had a pathological partial response to NAC. Herein, we report a rare case of curative surgery after NAC with FOLFOX for LACC in the sigmoid colon with extensive invasion of the abdominal wall. We reconstructed the extensive abdominal wall defect with a free anterolateral thigh flap. One of the optional treatment strategies for LACC with extensive abdominal wall invasion was reported in our report.
Collapse
Affiliation(s)
| | | | - Seigo Yukisawa
- Department of Medical Oncology, Saiseikai Utsunomiya Hospital, Tochigi, JPN
| | - Masaru Kimata
- Department of Surgery, Saiseikai Utsunomiya Hospital, Tochigi, JPN
| | | |
Collapse
|
3
|
Kataoka K, Yamada T, Yamazaki K, Mori K, Matsuhashi N, Shiozawa M, Iwai T, Goto M, Yasui M, Takii Y, Suto T, Takamizawa Y, Takase N, Sharma S, Ensor J, Jurdi A, Liu MC, Ikeda M, Kanemitsu Y. Trial Protocol of a Phase II Study of mFOLFOXIRI after Metastasectomy in Patients with Oligometastatic Colorectal Cancer (FANTASTIC Study). J Anus Rectum Colon 2024; 8:246-252. [PMID: 39086881 PMCID: PMC11286365 DOI: 10.23922/jarc.2024-025] [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: 03/21/2024] [Accepted: 04/23/2024] [Indexed: 08/02/2024] Open
Abstract
Background The survival benefit of adjuvant chemotherapy after surgical resection of oligometastases from colorectal cancer (CRC) remains unclear. The prognostic role of circulating-tumor DNA (ctDNA) was reported recently and a risk stratification strategy based on monitoring minimal/molecular residual disease (MRD) has been proposed, however, which drug regimen is most effective for ctDNA-positive patients is unknown. Methods/Design Oligometastatic CRC patients planning to undergo surgery were registered in this study. After metastasectomy, the registered patients were enrolled in the treatment arm, in which 8 courses of modified-FOLFOXIRI (mFOLFOXIRI; irinotecan 150 mg/m2, oxaliplatin 85 mg/m2, l-leucovorin (l-LV) 200 mg/m2, and 46-h continuous infusion of 5-fluorouracil (5-FU) 2400 mg/m2 every 2 weeks) followed by 4 courses of 5-FU/l-LV are administered. The patients who did not meet the eligibility criteria for the treatment arm or did not consent to mFOLFOXIRI enrolled in the observation arm in which standard of care treatment is provided. Prospective blood collections for retrospective ctDNA analysis are scheduled pre-surgery, and at 28 days, 4 and 7 months after surgery. The primary endpoint is treatment compliance at 8 courses of mFOLFOXIRI and the key secondary endpoints are the ctDNA-positivity rate and survival outcomes in ctDNA-positive and -negative groups. A total of 85 patients will be enrolled from 11 institutions. First patient-in was on July 2020. Accrual completed in February 2024. Discussion This study will potentially identify a better treatment strategy for patients with resectable oligometastatic CRC having postsurgical ctDNA positivity, compared to the current standard of care approaches.
Collapse
Affiliation(s)
- Kozo Kataoka
- Division of Lower GI, Department of Gastroenterological Surgery, Hyogo Medical University, Nishinomiya, Japan
| | - Takeshi Yamada
- Department of Gastrointestinal Surgery, Nippon Medical School, Tokyo, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shunto, Japan
| | - Keita Mori
- Department of Biostatistics, Clinical Research Center, Shizuoka Cancer Center, Shunto, Japan
| | - Nobuhisa Matsuhashi
- Department of Gastroenterological Surgery・Pediatric Surgery, Gifu University School of Medicine, Gifu, Japan
| | - Manabu Shiozawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Kanagawa, Japan
| | - Takuma Iwai
- Department of Gastrointestinal Surgery, Nippon Medical School, Tokyo, Japan
| | - Masahiro Goto
- The Second Department of Internal Medicine, Osaka Medical College Hospital, Takatsuki, Japan
| | - Masayoshi Yasui
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Yasumasa Takii
- Department of Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Takeshi Suto
- Department of Gastrointestinal Surgery, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Yasuyuki Takamizawa
- Department of Colorectal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Naoto Takase
- Department of Medical Oncology, Takarazuka City Hospital, Takarazuka, Japan
| | | | | | | | | | - Masataka Ikeda
- Division of Lower GI, Department of Gastroenterological Surgery, Hyogo Medical University, Nishinomiya, Japan
| | - Yukihide Kanemitsu
- Department of Colorectal Surgery, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
4
|
Ochiai K, Bhutiani N, Ikeda A, Uppal A, White MG, Peacock O, Messick CA, Bednarski BK, You YQN, Skibber JM, Chang GJ, Konishi T. Total Neoadjuvant Therapy for Rectal Cancer: Which Regimens to Use? Cancers (Basel) 2024; 16:2093. [PMID: 38893212 PMCID: PMC11171181 DOI: 10.3390/cancers16112093] [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: 04/11/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Total neoadjuvant therapy (TNT) is a novel strategy for rectal cancer that administers both (chemo)radiotherapy and systemic chemotherapy before surgery. TNT is expected to improve treatment compliance, tumor regression, organ preservation, and oncologic outcomes. Multiple TNT regimens are currently available with various combinations of the treatments including induction or consolidation chemotherapy, triplet or doublet chemotherapy, and long-course chemoradiotherapy or short-course radiotherapy. Evidence on TNT is rapidly evolving with new data on clinical trials, and no definitive consensus has been established on which regimens to use for improving outcomes. Clinicians need to understand the advantages and limitations of the available regimens for multidisciplinary decision making. This article reviews currently available evidence on TNT for rectal cancer. A decision making flow chart is provided for tailor-made use of TNT regimens based on tumor location and local and systemic risk.
Collapse
Affiliation(s)
- Kentaro Ochiai
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
- Department of Colon and Rectal Surgery, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Neal Bhutiani
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
| | - Atsushi Ikeda
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
| | - Abhineet Uppal
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
| | - Michael G. White
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
| | - Oliver Peacock
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
| | - Craig A. Messick
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
| | - Brian K. Bednarski
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
| | - Yi-Qian Nancy You
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
| | - John M. Skibber
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
| | - George J. Chang
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
| | - Tsuyoshi Konishi
- Department of Colon and Rectal Surgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (K.O.); (N.B.); (A.I.); (A.U.); (O.P.); (C.A.M.); (B.K.B.); (Y.-Q.N.Y.); (J.M.S.); (G.J.C.)
| |
Collapse
|
5
|
Lecomte T, Tougeron D, Chautard R, Bressand D, Bibeau F, Blanc B, Cohen R, Jacques J, Lagasse JP, Laurent-Puig P, Lepage C, Lucidarme O, Martin-Babau J, Panis Y, Portales F, Taieb J, Aparicio T, Bouché O. Non-metastatic colon cancer: French Intergroup Clinical Practice Guidelines for diagnosis, treatments, and follow-up (TNCD, SNFGE, FFCD, GERCOR, UNICANCER, SFCD, SFED, SFRO, ACHBT, SFP, AFEF, and SFR). Dig Liver Dis 2024; 56:756-769. [PMID: 38383162 DOI: 10.1016/j.dld.2024.01.208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 01/25/2024] [Accepted: 01/28/2024] [Indexed: 02/23/2024]
Abstract
INTRODUCTION This article is a summary of the French intergroup guidelines regarding the management of non-metastatic colon cancer (CC), revised in November 2022. METHODS These guidelines represent collaborative work of all French medical and surgical societies involved in the management of CC. Recommendations were graded in three categories (A, B, and C) according to the level of evidence found in the literature published up to November 2022. RESULTS Initial evaluation of CC is based on clinical examination, colonoscopy, chest-abdomen-pelvis computed tomography (CT) scan, and carcinoembryonic antigen (CEA) assay. CC is usually managed by surgery and adjuvant treatment depending on the pathological findings. The use of adjuvant therapy remains a challenging question in stage II disease. For high-risk stage II CC, adjuvant chemotherapy must be discussed and fluoropyrimidine monotherapy or oxaliplatin-based chemotherapy proposed according to the type and number of poor prognostic features. Oxaliplatin-based chemotherapy (FOLFOX or CAPOX) is the current standard for adjuvant therapy of patients with stage III CC. However, these regimens are associated with significant oxaliplatin-induced neurotoxicity. The results of the recent IDEA study provide evidence that 3 months of treatment with CAPOX is as effective as 6 months of oxaliplatin-based therapy in patients with low-risk stage III CC (T1-3 and N1). A 6-month oxaliplatin-based therapy remains the standard of care for high-risk stage III CC (T4 and/or N2). For patients unfit for oxaliplatin, fluoropyrimidine monotherapy is recommended. CONCLUSION French guidelines for non-metastatic CC management help to offer the best personalized therapeutic strategy in daily clinical practice. Each individual case must be discussed within a multidisciplinary tumor board and then the treatment option decided with the patient.
Collapse
Affiliation(s)
- Thierry Lecomte
- Department of Hepatogastroenterology and Digestive Oncology, Tours University Hospital, Tours, France; Inserm UMR 1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France.
| | - David Tougeron
- Department of Hepatogastroenterology, Poitiers University Hospital, Poitiers, France
| | - Romain Chautard
- Department of Hepatogastroenterology and Digestive Oncology, Tours University Hospital, Tours, France; Inserm UMR 1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | - Diane Bressand
- Department of Hepatogastroenterology and Digestive Oncology, Tours University Hospital, Tours, France
| | - Frédéric Bibeau
- Department of Pathology, Besançon University Hospital, Besançon, France
| | - Benjamin Blanc
- Department of Digestive Surgery, Dax Hospital, Dax, France
| | - Romain Cohen
- Sorbonne Université, Department of Medical Oncology, Saint-Antoine hospital, AP-HP, Inserm, Unité Mixte de Recherche Scientifique 938 et SiRIC CURAMUS, Saint-Antoine Research Center, Paris, France
| | - Jérémie Jacques
- Department of Hepatogastroenterology, Limoges University Hospital, Limoges, France
| | - Jean-Paul Lagasse
- Department of Hepatogastroenterology and Digestive Oncology, Orléans University Hospital, Orléans, France
| | - Pierre Laurent-Puig
- Department of Biology, AP-HP, European Georges Pompidou Hospital, Paris, France
| | - Come Lepage
- Department of Hepatogastroenterology and Digestive Oncology, Dijon University Hospital, Dijon, France
| | - Olivier Lucidarme
- Department of Radiology, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Jérôme Martin-Babau
- Armoricain Center of Radiotherapy, Radiology and Oncology, Côtes D'Armor Private Hospital, Plérin, France
| | - Yves Panis
- Department of Colorectal Surgery, AP-HP, Beaujon Hospital, Clichy, France
| | - Fabienne Portales
- Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, Paris, France
| | - Thomas Aparicio
- Department of Gastroenterology and Digestive Oncology, AP-HP, Saint-Louis Hospital, Paris, France
| | - Olivier Bouché
- Department of Digestive Oncology, Reims, CHU Reims, France
| |
Collapse
|
6
|
Lee KH, Yang IJ, Ha GW, Lee J, Park YY, Lee SH, Lee JM, Bae JH, Park EJ, Kim H, Kim KY, An S, Kim IY, Kim JY. mFOLFIRINOX versus mFOLFOX 6 as adjuvant treatment for high-risk stage III colon cancer - the FROST trial: study protocol for a multicenter, randomized controlled, phase II trial. BMC Cancer 2024; 24:397. [PMID: 38553680 PMCID: PMC10979582 DOI: 10.1186/s12885-024-11939-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 01/31/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND High-risk stage III colon cancer has a considerably poorer prognosis than stage II and low-risk stage III colon cancers. Nevertheless, most guidelines recommend similar adjuvant treatment approaches for all these stages despite the dearth of research focusing on high-risk stage III colon cancer and the potential for improved prognosis with intensive adjuvant treatment. Given the the proven efficacy of triplet chemotherapy in metastatic colorectal cancer treatment, the goal of this study is to evaluate the oncologic efficacy and safety of mFOLFIRINOX in comparison to those of the current standard of care, mFOLFOX 6, as an adjuvant treatment for patients diagnosed with high-risk stage III colon cancer after radical resection. METHODS This multicenter, randomized (1:1), open-label, phase II trial will assess and compare the effectiveness and toxicity of mFOLFIRINOX and mFOLFOX 6 in patients with high-risk stage III colon cancer after radical resection. The goal of the trial is to enroll 312 eligible patients, from 11 institutes, aged between 20 and 70 years, with an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2, or between 70 and 75 with an ECOG performance status of 0. Patients will be randomized into two arms - Arm A, the experimental arm, and Arm B, the reference arm - and will receive 12 cycles of mFOLFIRINOX and mFOLFOX 6 every 2 weeks, respectively. The primary endpoint of this study is the 3-year disease-free survival, and secondary endpoints include the 3-year overall survival and treatment toxicity. DISCUSSION The Frost trial would help determine the oncologic efficacy and safety of adjuvant triplet chemotherapy for high-risk stage III colon cancers and ultimately improve prognoses. TRIAL REGISTRATION ClinicalTrials.gov NCT05179889, registered on 17 December 2021.
Collapse
Affiliation(s)
- Kyung-Ha Lee
- Department of Colorectal Surgery, Chungnam National University Hospital & College of Medicine, Daejeon, Korea
| | - In Jun Yang
- Department of Colorectal Surgery, Chungnam National University Hospital & College of Medicine, Daejeon, Korea
| | - Gi Won Ha
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Korea
| | - Jaeim Lee
- Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
| | - Youn Young Park
- Department of Surgery, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Korea
| | - Suk Hwan Lee
- Department of Surgery, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Korea
| | - Jong Min Lee
- Department of Surgery, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Jung Hoon Bae
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun Jung Park
- Division of Colorectal Surgery, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyungjin Kim
- Department of Surgery, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Keun Young Kim
- Department of General Surgery, Wonkwang University Hospital & School of Medicine, Iksan, Korea
| | - Sanghyung An
- Department of Surgery, Yonsei University, Wonju College of Medicine, Wonju, Korea
| | - Ik Yong Kim
- Department of Surgery, Yonsei University, Wonju College of Medicine, Wonju, Korea
| | - Ji Yeon Kim
- Department of Colorectal Surgery, Chungnam National University Hospital & College of Medicine, Daejeon, Korea.
| |
Collapse
|
7
|
He L, Li H, Wang Y, Li W, Gao L, Xu B, Hu J, He P, Pu W, Sun G, Wang Z, Han Q, Liu B, Chen H. Complete remission in a pretreated, microsatellite-stable, KRAS-mutated colon cancer patient after treatment with sintilimab and bevacizumab and platinum-based chemotherapy: a case report and literature review. Front Immunol 2024; 15:1354613. [PMID: 38617840 PMCID: PMC11010642 DOI: 10.3389/fimmu.2024.1354613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/01/2024] [Indexed: 04/16/2024] Open
Abstract
Metastatic colon cancer remains an incurable disease, and it is difficult for existing treatments to achieve the desired clinical outcome, especially for colon cancer patients who have received first-line treatment. Although immune checkpoint inhibitors (ICIs) have demonstrated durable clinical efficacy in a variety of solid tumors, their response requires an inflammatory tumor microenvironment. However, microsatellite-stable (MSS) colon cancer, which accounts for the majority of colorectal cancers, is a cold tumor that does not respond well to ICIs. Combination regimens open the door to the utility of ICIs in cold tumors. Although combination therapies have shown their advantage even for MSS colon cancer, it remains unclear whether combination therapies show their advantage in patients with pretreated metastatic colon cancer. We report a patient who has achieved complete remission and good tolerance with sintilimab plus bevacizumab and platinum-based chemotherapy after postoperative recurrence. The patient had KRAS mutation and MSS-type colon cancer, and his PD-1+CD8+ and CD3-CD19-CD14+CD16-HLA-DR were both positive. He has achieved a progression-free survival of 43 months and is still being followed up at our center. The above results suggest that this therapeutic regimen is a promising treatment modality for the management of pretreated, MSS-type and KRAS-mutated metastatic colorectal cancer although its application to the general public still needs to be validated in clinical trials.
Collapse
Affiliation(s)
- Lijuan He
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Haiyuan Li
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yunpeng Wang
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Weidong Li
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lei Gao
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Bo Xu
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jike Hu
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Puyi He
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Weigao Pu
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Guodong Sun
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhuanfang Wang
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Qinying Han
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Ben Liu
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Hao Chen
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Provincial Key Laboratory Of Environmental Oncology, Lanzhou, China
| |
Collapse
|
8
|
Liu B, Polack M, Coudray N, Quiros AC, Sakellaropoulos T, Crobach AS, van Krieken JHJ, Yuan K, Tollenaar RA, Mesker WE, Tsirigos A. Self-Supervised Learning Reveals Clinically Relevant Histomorphological Patterns for Therapeutic Strategies in Colon Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582106. [PMID: 38496571 PMCID: PMC10942268 DOI: 10.1101/2024.02.26.582106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Self-supervised learning (SSL) automates the extraction and interpretation of histopathology features on unannotated hematoxylin-and-eosin-stained whole-slide images (WSIs). We trained an SSL Barlow Twins-encoder on 435 TCGA colon adenocarcinoma WSIs to extract features from small image patches. Leiden community detection then grouped tiles into histomorphological phenotype clusters (HPCs). HPC reproducibility and predictive ability for overall survival was confirmed in an independent clinical trial cohort (N=1213 WSIs). This unbiased atlas resulted in 47 HPCs displaying unique and sharing clinically significant histomorphological traits, highlighting tissue type, quantity, and architecture, especially in the context of tumor stroma. Through in-depth analysis of these HPCs, including immune landscape and gene set enrichment analysis, and association to clinical outcomes, we shed light on the factors influencing survival and responses to treatments like standard adjuvant chemotherapy and experimental therapies. Further exploration of HPCs may unveil new insights and aid decision-making and personalized treatments for colon cancer patients.
Collapse
Affiliation(s)
- Bojing Liu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Sweden
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, New York, USA
| | - Meaghan Polack
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Nicolas Coudray
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, New York, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| | | | - Theodore Sakellaropoulos
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, New York, USA
| | | | | | - Ke Yuan
- Department of Computing Science, University of Glasgow, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Rob A.E.M. Tollenaar
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Wilma E. Mesker
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, New York, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
9
|
Wijler LA, Viergever BJ, Strating E, van Schelven SJ, Poghosyan S, Frenkel NC, Te Rietmole H, Verheem A, Raats DAE, Borel Rinkes IHM, Hagendoorn J, Kranenburg O. Onward Spread from Liver Metastases Is a Major Cause of Multi-Organ Metastasis in a Mouse Model of Metastatic Colon Cancer. Cancers (Basel) 2024; 16:1073. [PMID: 38473429 DOI: 10.3390/cancers16051073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/15/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Colorectal cancer metastasizes predominantly to the liver but also to the lungs and the peritoneum. The presence of extra-hepatic metastases limits curative (surgical) treatment options and is associated with very poor survival. The mechanisms governing multi-organ metastasis formation are incompletely understood. Here, we tested the hypothesis that the site of tumor growth influences extra-hepatic metastasis formation. To this end, we implanted murine colon cancer organoids into the primary tumor site (i.e., the caecum) and into the primary metastasis site (i.e., the liver) in immunocompetent mice. The organoid-initiated liver tumors were significantly more efficient in seeding distant metastases compared to tumors of the same origin growing in the caecum (intra-hepatic: 51 vs. 40%, p = 0.001; peritoneal cavity: 51% vs. 33%, p = 0.001; lungs: 30% vs. 7%, p = 0.017). The enhanced metastatic capacity of the liver tumors was associated with the formation of 'hotspots' of vitronectin-positive blood vessels surrounded by macrophages. RNA sequencing analysis of clinical samples showed a high expression of vitronectin in liver metastases, along with signatures reflecting hypoxia, angiogenesis, coagulation, and macrophages. We conclude that 'onward spread' from liver metastases is facilitated by liver-specific microenvironmental signals that cause the formation of macrophage-associated vascular hotspots. The therapeutic targeting of these signals may help to contain the disease within the liver and prevent onward spread.
Collapse
Affiliation(s)
- Liza A Wijler
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Bastiaan J Viergever
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Esther Strating
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Susanne J van Schelven
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Susanna Poghosyan
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Nicola C Frenkel
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Hedy Te Rietmole
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Andre Verheem
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Danielle A E Raats
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Inne H M Borel Rinkes
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Jeroen Hagendoorn
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Onno Kranenburg
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Utrecht Platform for Organoid Technology, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
10
|
Pinato DJ, Fulgenzi CAM, D'Alessio A. Immunotherapy at all stages of hepatocellular carcinoma. Nat Med 2024; 30:640-641. [PMID: 38409592 DOI: 10.1038/s41591-024-02828-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Affiliation(s)
- David J Pinato
- Department of Surgery and Cancer, Hammersmith Hospital Campus, Imperial College London, London, UK.
- Department of Translational Medicine, Università Del Piemonte Orientale "A. Avogadro", Novara, Italy.
| | - Claudia A M Fulgenzi
- Department of Surgery and Cancer, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - Antonio D'Alessio
- Department of Surgery and Cancer, Hammersmith Hospital Campus, Imperial College London, London, UK
- Department of Translational Medicine, Università Del Piemonte Orientale "A. Avogadro", Novara, Italy
| |
Collapse
|
11
|
Pollini T, Tran T, Wong P, Adam MA, Alseidi A, Corvera C, Hirose K, Nakakura E, Warren R, Maker VK, Maker AV. Improved survival of patients receiving immunotherapy and chemotherapy following curative-intent resection of colorectal liver metastases. J Gastrointest Surg 2024; 28:246-251. [PMID: 38445916 DOI: 10.1016/j.gassur.2023.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 12/25/2023] [Indexed: 03/07/2024]
Abstract
BACKGROUND Despite significant advancements in the treatment of patients with colorectal liver metastases (CRLMs), only a minority will experience long-term survival. This study aimed to determine the effect of chemotherapy (CT) and immunotherapy (IT) compared with that of CT alone on patient survival after surgical resection. METHODS Patients undergoing curative-intent liver resection followed by adjuvant systemic therapy for stage IV colon cancer were identified using the National Cancer Database. Patients were stratified into type of therapy (CT alone vs CT + IT) and microsatellite status. Propensity score-weighted analysis was performed through 1:1 matching based on the nearest neighbor method. RESULTS Of 9943 patients who underwent resection of CRLMs, 7971 (80%) received systemic adjuvant therapy. Of 7971 patients, 1432 (18%) received a combination of CT and IT. Microsatellite status was not associated with overall survival (OS). Adjuvant CT + IT was associated with increased 3-year OS compared with that of CT alone in both the unmatched cohort (55% vs 48%, respectively; P < .001) and matched cohort (52% vs 48%, respectively; P = .050). On multivariate analysis, older age, positive resection margins, and KRAS mutation were independent predictors of poor survival, whereas the administration of adjuvant CT + IT was an independent predictor of improved survival. CONCLUSION IT combined with CT was associated with improved survival compared with that of CT alone after curative-intent resection of CRLMs, regardless of microsatellite instability status. Clinical trials to determine optimal patient selection, IT regimen, and long-term efficacy to improve outcomes of patients with CRLMs are warranted.
Collapse
Affiliation(s)
- Tommaso Pollini
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Thuy Tran
- Division of Surgical Oncology, Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, California, United States; Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Paul Wong
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Mohamed A Adam
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Adnan Alseidi
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Carlos Corvera
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Kenzo Hirose
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Eric Nakakura
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Robert Warren
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Vijay K Maker
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Ajay V Maker
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States.
| |
Collapse
|
12
|
Bao X, Li Q, Chen D, Dai X, Liu C, Tian W, Zhang H, Jin Y, Wang Y, Cheng J, Lai C, Ye C, Xin S, Li X, Su G, Ding Y, Xiong Y, Xie J, Tano V, Wang Y, Fu W, Deng S, Fang W, Sheng J, Ruan J, Zhao P. A multiomics analysis-assisted deep learning model identifies a macrophage-oriented module as a potential therapeutic target in colorectal cancer. Cell Rep Med 2024; 5:101399. [PMID: 38307032 PMCID: PMC10897549 DOI: 10.1016/j.xcrm.2024.101399] [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: 03/06/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 02/04/2024]
Abstract
Colorectal cancer (CRC) is a common malignancy involving multiple cellular components. The CRC tumor microenvironment (TME) has been characterized well at single-cell resolution. However, a spatial interaction map of the CRC TME is still elusive. Here, we integrate multiomics analyses and establish a spatial interaction map to improve the prognosis, prediction, and therapeutic development for CRC. We construct a CRC immune module (CCIM) that comprises FOLR2+ macrophages, exhausted CD8+ T cells, tolerant CD8+ T cells, exhausted CD4+ T cells, and regulatory T cells. Multiplex immunohistochemistry is performed to depict the CCIM. Based on this, we utilize advanced deep learning technology to establish a spatial interaction map and predict chemotherapy response. CCIM-Net is constructed, which demonstrates good predictive performance for chemotherapy response in both the training and testing cohorts. Lastly, targeting FOLR2+ macrophage therapeutics is used to disrupt the immunosuppressive CCIM and enhance the chemotherapy response in vivo.
Collapse
Affiliation(s)
- Xuanwen Bao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China.
| | - Qiong Li
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Dong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Xiaomeng Dai
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Chuan Liu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Weihong Tian
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hangyu Zhang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Yuzhi Jin
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Yin Wang
- College of Computer Science and Technology, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Jinlin Cheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Chunyu Lai
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Chanqi Ye
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Shan Xin
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Xin Li
- Department of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ge Su
- College of Computer Science and Technology, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Yongfeng Ding
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Yangyang Xiong
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Jindong Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Vincent Tano
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637551, Republic of Singapore
| | - Yanfang Wang
- Ludwig-Maximilians-Universität München (LMU), 80539 Munich, Germany
| | - Wenguang Fu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Shuiguang Deng
- College of Computer Science and Technology, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Jianpeng Sheng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China.
| | - Jian Ruan
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China; Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China.
| |
Collapse
|
13
|
Audisio A, Fazio R, Daprà V, Assaf I, Hendlisz A, Sclafani F. Neoadjuvant chemotherapy for early-stage colon cancer. Cancer Treat Rev 2024; 123:102676. [PMID: 38160535 DOI: 10.1016/j.ctrv.2023.102676] [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: 10/31/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Surgery with or without adjuvant chemotherapy is the standard treatment for early-stage colon cancer. However, evidence has recently emerged for neoadjuvant chemotherapy, with the results of randomised clinical trials sparking debates within multidisciplinary teams and splitting the gastrointestinal oncology community. Further to a systematic search of the literature, we provide a thorough and in-depth analysis of the findings from these trials, highlighting the advantages and disadvantages of neoadjuvant chemotherapy. We conclude that, while there is a potential value of moving systemic therapy from the post-operative to the pre-operative setting, the available evidence does not justify a shift in the treatment paradigm of early-stage colon cancer, and surgery with or without adjuvant chemotherapy should remain the standard approach for these patients.
Collapse
Affiliation(s)
- Alessandro Audisio
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium
| | - Roberta Fazio
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium
| | - Valentina Daprà
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium
| | - Irene Assaf
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium; Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alain Hendlisz
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium; Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Francesco Sclafani
- Institut Jules Bordet, The Brussels University Hospital (HUB), Brussels, Belgium; Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
14
|
Underwood PW, Ruff SM, Pawlik TM. Update on Targeted Therapy and Immunotherapy for Metastatic Colorectal Cancer. Cells 2024; 13:245. [PMID: 38334637 PMCID: PMC10854977 DOI: 10.3390/cells13030245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/21/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Metastatic colorectal cancer remains a deadly malignancy and is the third leading cause of cancer-related death. The mainstay of treatment for metastatic colorectal cancer is chemotherapy, but unfortunately, even with recent progress, overall survival is still poor. Colorectal cancer is a heterogeneous disease, and the underlying genetic differences among tumors can define the behavior and prognosis of the disease. Given the limitations of cytotoxic chemotherapy, research has focused on developing targeted therapy based on molecular subtyping. Since the early 2000s, multiple targeted therapies have demonstrated efficacy in treating metastatic colorectal cancer and have received FDA approval. The epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), and DNA mismatch repair pathways have demonstrated promising results for targeted therapies. As new gene mutations and proteins involved in the oncogenesis of metastatic colorectal cancer are identified, new targets will continue to emerge. We herein provide a summary of the updated literature regarding targeted therapies for patients with mCRC.
Collapse
Affiliation(s)
| | | | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, 395 W. 12th Ave., Suite 670, Columbus, OH 43210, USA; (P.W.U.); (S.M.R.)
| |
Collapse
|
15
|
D'Alessio A, Rimassa L. The long and winding road: Adjuvant therapy for early-stage hepatocellular carcinoma. MED 2024; 5:7-9. [PMID: 38218177 DOI: 10.1016/j.medj.2023.11.004] [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: 11/06/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 01/15/2024]
Abstract
Despite being the first positive phase 3 trial in the adjuvant setting for early-stage hepatocellular carcinoma, the IMbrave050 study raises a number of questions regarding patient selection, endpoint robustness, and the balance between efficacy and acceptable toxicity.1.
Collapse
Affiliation(s)
- Antonio D'Alessio
- Department of Surgery & Cancer, Imperial College London, London, UK; Department of Translational Medicine, University of Piemonte Orientale, Novara, 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.
| |
Collapse
|
16
|
Trembath HE, Yeh JJ, Lopez NE. Gastrointestinal Malignancy: Genetic Implications to Clinical Applications. Cancer Treat Res 2024; 192:305-418. [PMID: 39212927 DOI: 10.1007/978-3-031-61238-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Advances in molecular genetics have revolutionized our understanding of the pathogenesis, progression, and therapeutic options for treating gastrointestinal (GI) cancers. This chapter provides a comprehensive overview of the molecular landscape of GI cancers, focusing on key genetic alterations implicated in tumorigenesis across various anatomical sites including GIST, colon and rectum, and pancreas. Emphasis is placed on critical oncogenic pathways, such as mutations in tumor suppressor genes, oncogenes, chromosomal instability, microsatellite instability, and epigenetic modifications. The role of molecular biomarkers in predicting prognosis, guiding treatment decisions, and monitoring therapeutic response is discussed, highlighting the integration of genomic profiling into clinical practice. Finally, we address the evolving landscape of precision oncology in GI cancers, considering targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Hannah E Trembath
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Jen Jen Yeh
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Nicole E Lopez
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA.
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA.
| |
Collapse
|
17
|
Yokoi R, Tajima JY, Fukada M, Hayashi H, Kuno M, Asai R, Sato Y, Yasufuku I, Kiyama S, Tanaka Y, Murase K, Matsuhashi N. Optimizing Treatment Strategy for Oligometastases/Oligo-Recurrence of Colorectal Cancer. Cancers (Basel) 2023; 16:142. [PMID: 38201569 PMCID: PMC10777959 DOI: 10.3390/cancers16010142] [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: 11/28/2023] [Revised: 12/25/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer, and nearly half of CRC patients experience metastases. Oligometastatic CRC represents a distinct clinical state characterized by limited metastatic involvement, demonstrating a less aggressive nature and potentially improved survival with multidisciplinary treatment. However, the varied clinical scenarios giving rise to oligometastases necessitate a precise definition, considering primary tumor status and oncological factors, to optimize treatment strategies. This review delineates the concepts of oligometastatic CRC, encompassing oligo-recurrence, where the primary tumor is under control, resulting in a more favorable prognosis. A comprehensive examination of multidisciplinary treatment with local treatments and systemic therapy is provided. The overarching objective in managing oligometastatic CRC is the complete eradication of metastases, offering prospects of a cure. Essential to this management approach are local treatments, with surgical resection serving as the standard of care. Percutaneous ablation and stereotactic body radiotherapy present less invasive alternatives for lesions unsuitable for surgery, demonstrating efficacy in select cases. Perioperative systemic therapy, aiming to control micrometastatic disease and enhance local treatment effectiveness, has shown improvements in progression-free survival through clinical trials. However, the extension of overall survival remains variable. The review emphasizes the need for further prospective trials to establish a cohesive definition and an optimized treatment strategy for oligometastatic CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Nobuhisa Matsuhashi
- Department of Gastroenterological Surgery and Pediatric Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu City 501-1194, Gifu, Japan; (R.Y.); (K.M.)
| |
Collapse
|
18
|
Pesántez D, Ten Hoorn S, Machado I, García-Albéniz X, Rodríguez-Salas N, Heredia-Soto V, Viñal D, Pericay C, García-Carbonero R, Losa F, Alonso V, Vera R, Feliu Batlle J, Gallego J, Salud A, Nogué M, Layos L, Montagut C, Capdevila J, Vermeulen L, Maurel J, Fernandez-Martos C. Total neoadjuvant therapy with or without aflibercept in rectal cancer: 3-year results of GEMCAD-1402. J Natl Cancer Inst 2023; 115:1497-1505. [PMID: 37405857 DOI: 10.1093/jnci/djad120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/25/2023] [Accepted: 06/10/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND The results of the Grupo Español Multidisciplinar en Cáncer Digestivo (GEMCAD)-1402 phase II randomized trial suggested that adding aflibercept to modified fluorouracil, oxaliplatin, and leucovorin (mFOLFOX6) induction, followed by chemoradiation and surgery, could increase the pathological complete response (pCR) rate in patients with high-risk, locally advanced rectal cancer. Here we update results up to 3 years of follow-up and evaluate the predictive value of consensus molecular subtypes identified with immunohistochemistry (IHC). METHODS Patients with magnetic resonance imaging-defined T3c-d and/or T4 and/or N2 rectal adenocarcinoma in the middle or distal third were randomly assigned to mFOLFOX6 induction, with aflibercept (mF+A; n = 115) or without aflibercept (mF; n = 65), followed by capecitabine plus radiotherapy and surgery. The risk local relapse, distant metastases, disease-free survival (DFS), and overall survival (OS) were estimated at 3 years. Selected samples were classified via IHC into immune-infiltrate, epithelial, or mesenchymal subtypes. RESULTS mF+A and mF had 3-year DFS of 75.2% (95% confidence interval [CI] = 66.1% to 82.2%) and 81.5% (95% CI = 69.8% to 89.1%), respectively; 3-year OS of 89.3% (95% CI = 82.0% to 93.8%) and 90.7% (95% CI = 80.6% to 95.7%), respectively; 3-year cumulative local relapse incidences of 5.2% (95% CI = 1.9% to 11.0%) and 6.1% (95% CI = 1.7% to 15.0%), respectively; and 3-year cumulative distant metastases rates of 17.3% (95% CI = 10.9% to 25.5%) and 16.9% (95% CI = 8.7% to 28.2%), respectively. pCRs were achieved in 27.5% (n = 22 of 80) and 0% (n = 0 of 10) of patients with epithelial and mesenchymal subtypes, respectively. CONCLUSION Adding aflibercept to mFOLFOX6 induction was not associated with improved DFS or OS. Our findings suggested that consensus molecular subtypes identified with IHC subtypes could be predictive of pCR with this treatment.
Collapse
Affiliation(s)
- David Pesántez
- Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Sanne Ten Hoorn
- Department of Medical Oncology, Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
- Department of Medical Oncology, Oncode Institute, Amsterdam, the Netherlands
| | - Isidro Machado
- Department of Pathology, Instituto Valenciano de Oncologia and Pathology Department, Hospital Quirón Salud, Valencia, Spain
| | | | - Nuria Rodríguez-Salas
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain
- Translational Oncology Group, IdiPAZ, Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Medical Oncology, Centro de Investigación Biomédica en Red - Cáncer (CIBERONC), Madrid, Spain
| | - Victoria Heredia-Soto
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain
- Translational Oncology Group, IdiPAZ, Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Medical Oncology, Centro de Investigación Biomédica en Red - Cáncer (CIBERONC), Madrid, Spain
| | - David Viñal
- Department of Medical Oncology, Complex Sanitari Parc Tauli, Sabadell, Spain
| | - Carles Pericay
- Department of Medical Oncology, Complex Sanitari Parc Tauli, Sabadell, Spain
| | | | - Ferran Losa
- Medical Oncology Department, Hospital Sant Joan Despí - Moises Broggi, Barcelona, Spain
| | - Vicente Alonso
- Medical Oncology Department, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Ruth Vera
- Medical Oncology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Jaime Feliu Batlle
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain
- Translational Oncology Group, IdiPAZ, Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Medical Oncology, Centro de Investigación Biomédica en Red - Cáncer (CIBERONC), Madrid, Spain
| | - Javier Gallego
- Medical Oncology Department, Hospital Universitario de Alicante, Alicante, Spain
| | - Antonieta Salud
- Medical Oncology Department, Hospital Universitario Arnau de Vilanova, Lleida, Spain
| | - Miquel Nogué
- Medical Oncology Department, Hospital de Granollers, Barcelona, Spain
| | - Laura Layos
- Medical Oncology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Clara Montagut
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Jaume Capdevila
- Medical Oncology Department, Vall Hebron University Hospital, Vall Hebron Institute of Oncology, IOB Quiron-Teknon, Barcelona, Spain
| | - Louis Vermeulen
- Department of Medical Oncology, Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
- Department of Medical Oncology, Oncode Institute, Amsterdam, the Netherlands
| | - Joan Maurel
- Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Carlos Fernandez-Martos
- Department of Medical Oncology, Initia Oncology, Hospital Quirón Salud Valencia, Valencia, Spain
| |
Collapse
|
19
|
Chitkara A, Kaur N, Desai A, Mehta D, Anamika F, Sarkar S, Gowda N, Sethi P, Thawani R, Chen EY. Risks of hypertension and thromboembolism in patients receiving bevacizumab with chemotherapy for colorectal cancer: A systematic review and meta-analysis. Cancer Med 2023; 12:21579-21591. [PMID: 38069531 PMCID: PMC10757147 DOI: 10.1002/cam4.6662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/12/2023] [Accepted: 10/03/2023] [Indexed: 12/31/2023] Open
Abstract
BACKGROUND Guidelines show that for metastatic colorectal cancer (mCRC), a combination of three-drug regimens, fluorouracil, leucovorin, and oxaliplatin and bevacizumab (BVZ), is one of the first-line standard therapies. BVZ is generally well tolerated; however, it is associated with infrequent, life-threatening side effects such as severe hypertension (HTN) (5%-18%), Grade ≥3 arterial thromboembolism (ATE) (2.6%), Grade ≥3 hemorrhagic events (1.2%-4.6%), and gastrointestinal perforation (0.3%-2.4%). This meta-analysis aims to evaluate the additive risk of BVZ-induced severe HTN and thromboembolism when BVZ is combined with a standard chemotherapy regime in patients with mCRC. METHODS Our search was conducted from January 29, 2022, to February 22, 2022, through databases of PubMed, clinicaltrial.gov, EMBASE, Web of Science, and Cochrane Library. Data analysis from randomized controlled trials (RCTs) and clinical trials was conducted using Review Manager V.5.4, comparing BVZ-chemotherapy to chemotherapy only, focusing on cardiovascular AE such as HTN and arterial and venous thromboembolism. RESULTS The analysis from 26 clinical trials and RCTs showed that the odds of HTN were about four times higher, and ATE subgroup analysis of 11 studies showed over two times higher odds of ATE in patients being treated with BVZ compared to the chemotherapy-only group. CONCLUSION BVZ, when added to the standard chemotherapy regimen for mCRC, was associated with higher odds of developing HTN and thromboembolism, specifically ATE, than the chemotherapy-only group. Our findings are significant as they provide vital information in analyzing the risk-benefit ratio of adding BVZ to the standard chemotherapy regime in patients with mCRC, especially in patients with vascular comorbidities.
Collapse
Affiliation(s)
- Akshit Chitkara
- Internal MedicineUniversity of California RiversideRiversideCaliforniaUSA
| | - Nirmaljot Kaur
- Internal MedicineUniversity of California RiversideRiversideCaliforniaUSA
| | - Aditya Desai
- Internal MedicineUniversity of California RiversideRiversideCaliforniaUSA
| | - Devanshi Mehta
- Loma Linda UniversityCalifornia in Internal MedicineCaliforniaUSA
| | - Fnu Anamika
- Internal MedicineHackensack Meridian Ocean UniversityBrickNew JerseyUSA
| | - Srawani Sarkar
- Research LabAlbert Einstein College of MedicineNew YorkNew YorkUSA
| | - Nandini Gowda
- Internal MedicineUniversity of California RiversideRiversideCaliforniaUSA
| | - Prabhdeep Sethi
- Internal MedicineUniversity of California RiversideRiversideCaliforniaUSA
| | - Rajat Thawani
- Division of Hematology and Medical Oncology, Knight Cancer InstituteOregon Health & Sciences UniversityPortlandOregonUSA
| | - Emerson Y. Chen
- Division of Hematology and Medical Oncology, Knight Cancer InstituteOregon Health & Sciences UniversityPortlandOregonUSA
| |
Collapse
|
20
|
Ruff SM, Brown ZJ, Pawlik TM. A review of targeted therapy and immune checkpoint inhibitors for metastatic colorectal cancer. Surg Oncol 2023; 51:101993. [PMID: 37742544 DOI: 10.1016/j.suronc.2023.101993] [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: 06/02/2023] [Revised: 08/29/2023] [Accepted: 09/17/2023] [Indexed: 09/26/2023]
Abstract
Surgical resection is the cornerstone of treatment for metastatic colorectal cancer (CRC) and offers the best chance at long-term survival. Unfortunately, most patients do not present with resectable metastatic disease and, among patients who do undergo curative-intent resection, many will develop recurrence. In turn, patients require a multi-disciplinary treatment approach with a combination of chemotherapy, surgery, radiation, and/or liver directed therapies that is guided by patient disease burden and clinical status. The development of targeted therapies has led to varying success in other cancers and has emerged as a treatment option for patients with metastatic CRC. While cytotoxic chemotherapy aims to kill cells as they replicate, targeted therapies are directed at biologic features of cancers, like angiogenesis or immune checkpoints. Targeted therapy can facilitate a more treatment tailored approach to the unique genomic alterations of the tumor and hopefully deliver more personalized therapy. We herein provide a systematic review of approved targeted therapies and immune checkpoint inhibitors for metastatic CRC and provide an overview of the current literature.
Collapse
Affiliation(s)
- Samantha M Ruff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Zachary J Brown
- Department of Surgery, Division of Surgical Oncology, New York University Long Island, Mineola, NY, USA
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, 43210, USA.
| |
Collapse
|
21
|
Dianat-Moghadam H, Nedaeinia R, Keshavarz M, Azizi M, Kazemi M, Salehi R. Immunotherapies targeting tumor vasculature: challenges and opportunities. Front Immunol 2023; 14:1226360. [PMID: 37727791 PMCID: PMC10506263 DOI: 10.3389/fimmu.2023.1226360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/31/2023] [Indexed: 09/21/2023] Open
Abstract
Angiogenesis is a hallmark of cancer biology, and neoadjuvant therapies targeting either tumor vasculature or VEGF signaling have been developed to treat solid malignant tumors. However, these therapies induce complete vascular depletion leading to hypoxic niche, drug resistance, and tumor recurrence rate or leading to impaired delivery of chemo drugs and immune cell infiltration at the tumor site. Achieving a balance between oxygenation and tumor growth inhibition requires determining vascular normalization after treatment with a low dose of antiangiogenic agents. However, monotherapy within the approved antiangiogenic agents' benefits only some tumors and their efficacy improvement could be achieved using immunotherapy and emerging nanocarriers as a clinical tool to optimize subsequent therapeutic regimens and reduce the need for a high dosage of chemo agents. More importantly, combined immunotherapies and nano-based delivery systems can prolong the normalization window while providing the advantages to address the current treatment challenges within antiangiogenic agents. This review summarizes the approved therapies targeting tumor angiogenesis, highlights the challenges and limitations of current therapies, and discusses how vascular normalization, immunotherapies, and nanomedicine could introduce the theranostic potentials to improve tumor management in future clinical settings.
Collapse
Affiliation(s)
- Hassan Dianat-Moghadam
- Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mehdi Azizi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Kazemi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
22
|
Tan KF, In LLA, Vijayaraj Kumar P. Surface Functionalization of Gold Nanoparticles for Targeting the Tumor Microenvironment to Improve Antitumor Efficiency. ACS APPLIED BIO MATERIALS 2023; 6:2944-2981. [PMID: 37435615 DOI: 10.1021/acsabm.3c00202] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Gold nanoparticles (AuNPs) have undergone significant research for their use in the treatment of cancer. Numerous researchers have established their potent antitumor properties, which have greatly impacted the treatment of cancer. AuNPs have been used in four primary anticancer treatment modalities, namely radiation, photothermal therapy, photodynamic therapy, and chemotherapy. However, the ability of AuNPs to destroy cancer is lacking and can even harm healthy cells without the right direction to transport them to the tumor microenvironment. Consequently, a suitable targeting technique is needed. Based on the distinct features of the human tumor microenvironment, this review discusses four different targeting strategies that target the four key features of the tumor microenvironment, including abnormal vasculature, overexpression of specific receptors, an acidic microenvironment, and a hypoxic microenvironment, to direct surface-functionalized AuNPs to the tumor microenvironment and increase antitumor efficacies. In addition, some current completed or ongoing clinical trials of AuNPs will also be discussed below to further reinforce the concept of using AuNPs in anticancer therapy.
Collapse
Affiliation(s)
- Kin Fai Tan
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Palanirajan Vijayaraj Kumar
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
23
|
Huang S, Ye J, Gao X, Huang X, Huang J, Lu L, Lu C, Li Y, Luo M, Xie M, Lin Y, Liang R. Progress of research on molecular targeted therapies for colorectal cancer. Front Pharmacol 2023; 14:1160949. [PMID: 37614311 PMCID: PMC10443711 DOI: 10.3389/fphar.2023.1160949] [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: 02/07/2023] [Accepted: 07/26/2023] [Indexed: 08/25/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies, accounting for approximately 10% of global cancer incidence and mortality. Approximately 20% of patients with CRC present metastatic disease (mCRC) at the time of diagnosis. Moreover, up to 50% of patients with localized disease eventually metastasize. mCRC encompasses a complex cascade of reactions involving multiple factors and processes, leading to a diverse array of molecular mechanisms. Improved comprehension of the pathways underlying cancer cell development and proliferation, coupled with the accessibility of relevant targeted agents, has propelled advancements in CRC treatment, ultimately leading to enhanced survival rates. Mutations in various pathways and location of the primary tumor in CRC influences the efficacy of targeted agents. This review summarizes available targeted agents for different CRC pathways, with a focus on recent advances in anti-angiogenic and anti-epidermal growth factor receptor agents, BRAF mutations, and human epidermal growth factor receptor 2-associated targeted agents.
Collapse
Affiliation(s)
- Shilin Huang
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jiazhou Ye
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xing Gao
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xi Huang
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Julu Huang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lu Lu
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Cheng Lu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yongqiang Li
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Min Luo
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Mingzhi Xie
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yan Lin
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Rong Liang
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
24
|
Xu H, Li P, Ma H, Tan Y, Wang X, Cai F, Xu J, Sun H, Zhuang H, Hua Z. ADT-OH synergistically enhanced the antitumor activity of celecoxib in human colorectal cancer cells. Cancer Med 2023; 12:17193-17211. [PMID: 37492969 PMCID: PMC10501245 DOI: 10.1002/cam4.6342] [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: 04/07/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Colorectal cancer is one of the most prevalent cancers in the world, but the research on its prevention, early diagnosis and treatment is still a major challenge in clinical oncology. Thus, there is a pressing requirement to find effective strategies to improve the survival of colon cancer patients. METHODS Celecoxib has been accounted to be an effective antitumor drug, but may exhibit significant side effects. In recent studies, 5-(4-hydroxyphenyl)-3H-1,2-dithiole-3-thione (ADT-OH), one of the most commonly used reagents for the synthesis of sustained-release H2 S donors, has also been reported to inhibit cancer progression by affecting processes such as cell cycle, angiogenesis, and apoptosis. Therefore, we evaluated the therapeutic effect of the combination of ADT-OH and celecoxib on colorectal cancer through in vitro and in vivo, hoping to achieve better therapeutic effect and reduce the effect of celecoxib on gastric injury through exogenous administration of H2 S. RESULTS Our results demonstrated that ADT-OH combined with celecoxib synergistically inhibited the proliferation and migration ability of human colorectal cancer HCT116 cells, altered cell cycle and cytoskeleton, increased intracellular reactive oxygen species (ROS), and promoted cell apoptosis. Noteworthy, in vivo studies also indicated the excellent antitumor therapeutic effect of the combination therapy without apparent toxicity. CONCLUSIONS In general, our results provide a reasonable combination strategy of low-dose ADT-OH and celecoxib in the preclinical application of colorectal cancer.
Collapse
Affiliation(s)
- Huangru Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingP.R. China
| | - Ping Li
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingP.R. China
| | - Hailin Ma
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingP.R. China
| | - Yuanhao Tan
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingP.R. China
| | - Xiaoyang Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingP.R. China
| | - Fangfang Cai
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingP.R. China
- School of BiopharmacyChina Pharmaceutical UniversityNanjingChina
| | - Jiaqi Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingP.R. China
| | - Huisong Sun
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingP.R. China
| | - Hongqin Zhuang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingP.R. China
| | - Zi‐Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingP.R. China
- School of BiopharmacyChina Pharmaceutical UniversityNanjingChina
- Changzhou High‐Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc.ChangzhouP.R. China
| |
Collapse
|
25
|
Yang M, Yang C, Ma D, Li Z, Zhao W, Yang D. Single-cell analysis reveals cellular reprogramming in advanced colon cancer following FOLFOX-bevacizumab treatment. Front Oncol 2023; 13:1219642. [PMID: 37576892 PMCID: PMC10421721 DOI: 10.3389/fonc.2023.1219642] [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: 05/09/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction The combination of FOLFOX and bevacizumab (FOLFOX-Bev) is a promising treatment for advanced colorectal cancer (CRC). However, the response of the tumor microenvironment to FOLFOX-Bev is still largely unexplored. Methods We conducted single-cell transcriptomic analysis of CRC samples derived from a patient before and after treatment to gain insights into the cellular changes associated with FOLFOX-Bev treatment. Results We found that cancer cells with high proliferative, metastatic, and pro-angiogenic properties respond better to FOLFOX-Bev treatment. Moreover, FOLFOX-Bev enhances CD8+ T cell cytotoxicity, thereby boosting the anti-tumor immune response. Conversely, FOLFOX-Bev impairs the functionality of tumor-associated macrophages, plasma cells, and cancer-associated fibroblasts, leading to a decrease in VEGFB-mediated angiogenesis. Furthermore, FOLFOX-Bev treatment reset intercellular communication, which could potentially affect the function of non-cancer cells. Discussion Our findings provide valuable insights into the molecular mechanisms underlying the response of advanced CRC to FOLFOX-Bev treatment and highlight potential targets for improving the efficacy of this treatment strategy.
Collapse
Affiliation(s)
- Meiling Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ciqiu Yang
- Department of Breast Cancer, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Dong Ma
- Medical Oncology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zijun Li
- Guangdong Provincial Institute of Geriatrics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wei Zhao
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou, China
| | - Dongyang Yang
- Medical Oncology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Ruff SM, Shannon AH, Pawlik TM. The Role of Targeted Therapy in the Multi-Disciplinary Approach to Colorectal Liver Metastasis. Cancers (Basel) 2023; 15:3513. [PMID: 37444625 DOI: 10.3390/cancers15133513] [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: 05/26/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Colorectal cancer (CRC) is the second most common cause of cancer-related mortality in the United States. Among newly diagnosed patients with CRC, 20% will present with metastatic disease and another 25% will develop metastases. The surgical resection of the primary tumor and metastatic disease sites confers the best chance at long-term survival. Unfortunately, many patients will recur after resection or present with unresectable disease. As such, metastatic CRC is commonly treated with a combination of surgery, systemic therapy, and/or liver-directed therapies. Despite best efforts, 5-year survival for unresectable metastatic CRC is only about 20%. CRC is a heterogeneous disease and the underlying genetic differences inform behavior, treatment strategy, and prognosis. Given the limitations of cytotoxic chemotherapy and the growing role of molecular profiling, research has focused on identifying and developing targeted therapies. We herein review how genetic profiling informs prognosis, crucial cell-signaling pathways that play a role in CRC carcinogenesis, and currently approved targeted therapies for metastatic CRC.
Collapse
Affiliation(s)
- Samantha M Ruff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Alexander H Shannon
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
27
|
Murono K, Yokoyama Y, Nozawa H, Sasaki K, Emoto S, Matsuzaki H, Kashiwabara K, Ishigami H, Gohda Y, Yamaguchi H, Kitayama J, Ishihara S. Intraperitoneal paclitaxel combined with FOLFOX/CAPOX plus bevacizumab for colorectal cancer with peritoneal carcinomatosis (the iPac-02 trial): study protocol of a single arm, multicenter, phase 2 study. Int J Colorectal Dis 2023; 38:173. [PMID: 37340243 PMCID: PMC10282041 DOI: 10.1007/s00384-023-04434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND The safety of intraperitoneally administrated paclitaxel (op PTX) was demonstrated in the phase I trial of ip PTX combined with conventional systemic chemotherapy for colorectal cancer with peritoneal carcinomatosis. Moreover, the median survival time was 29.3 months, which was longer than that observed in previous studies. Here, we planned the phase II trial of ip PTX: the iPac-02 trial. METHODS This multicenter, open-label, single assignment interventional clinical study includes patients with colorectal cancer with unresectable peritoneal carcinomatosis. FOLFOX-bevacizumab or CAPOX-bevacizumab is administered concomitantly as systemic chemotherapy. PTX 20 mg/m2 is administered weekly through the peritoneal access port in addition to these conventional systemic chemotherapies. The response rate is the primary endpoint. Progression-free survival, overall survival, peritoneal cancer index improvement rate, rate of negative peritoneal lavage cytology, safety, and response rate to peritoneal metastases are the secondary endpoints. A total of 38 patients are included in the study. In the interim analysis, the study will continue to the second stage if at least 4 of the first 14 patients respond to the study treatment. The study has been registered at the Japan Registry of Clinical Trials (jRCT2031220110). RESULTS We previously conducted phase I trial of ip PTX combined with conventional systemic chemotherapy for colorectal cancer with peritoneal carcinomatosis [1]. In the study, three patients underwent mFOLFOX, bevacizumab, and weekly ip PTX, and the other three patients underwent CAPOX, bevacizumab, and weekly ip PTX treatment. The dose of PTX was 20 mg/m [2]. The primary endpoint was the safety of the chemotherapy, and secondary endpoints were response rate, peritoneal cancer index improvement rate, rate of negative peritoneal lavage cytology, progression-free survival, and overall survival. Dose limiting toxicity was not observed, and the adverse events of ip PTX combined with oxaliplatin-based systemic chemotherapy were similar to those described in previous studies using systemic chemotherapy alone [3, 4]. The response rate was 25%, peritoneal cancer index improvement rate was 50%, and cytology in peritoneal lavage turned negative in all the cases. The progression-free survival was 8.8 months (range, 6.8-12 months), and median survival time was 29.3 months [5], which was longer than that observed in previous studies. CONCLUSION Here, we planned the phase II trial of ip paclitaxel combined with conventional chemotherapy for colorectal cancer with peritoneal carcinomatosis: the iPac-02 trial.
Collapse
Affiliation(s)
- Koji Murono
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Yuichiro Yokoyama
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroaki Nozawa
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazuhito Sasaki
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shigenobu Emoto
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroyuki Matsuzaki
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kosuke Kashiwabara
- Interfaculty Initiative in Information Studies, Department of Biostatistics, School of Public Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hironori Ishigami
- Department of Chemotherapy, The University of Tokyo Hospital, Tokyo, Japan
| | - Yoshimasa Gohda
- Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hironori Yamaguchi
- Department of Clinical Oncology, Jichi Medical University Hospital, Shimotsuke, Japan
| | - Joji Kitayama
- Clinical Research Center, Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Soichiro Ishihara
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
28
|
Deng J, Zeng X, Hu W, Yue T, Luo Z, Zeng L, Li P, Chen J. Different doses of bevacizumab in combination with chemotherapy for advanced colorectal cancer: a meta-analysis and Bayesian analysis. Int J Colorectal Dis 2023; 38:164. [PMID: 37289304 DOI: 10.1007/s00384-023-04442-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/21/2023] [Indexed: 06/09/2023]
Abstract
OBJECTIVE The aim of the present study was to explore the incremental benefit of bevacizumab (Bev) in the treatment of advanced colorectal cancer (CRC) with different doses. METHODS A literature search of eight electronic databases (China National Knowledge Infrastructure, Wanfang databases, Chinese Biomedical Database, VIP medicine information, Cochrane Library, MEDLINE, PubMed, and EMBASE) was conducted from database creation to December 2022. Randomized controlled trials (RCTs) that compared Bev at various dosages + chemotherapy (CT) versus placebo (or blank control) + CT were selected. The overall survival (OS), progression-free survival (PFS), overall response rate (ORR; complete response [CR] + partial response [PR]), and grade ≥ 3 adverse events (AEs) were integrated first by pooled analysis. The likelihood of ideal dosage of Bev was then ranked using random effects within Bayesian analysis. RESULTS Twenty-six RCTs involving 18,261 patients met the inclusion criteria. OS increased significantly after using 5 mg (HR: 0.87, 95% CI 0.75 to 1.00) and 10 mg dosages of Bev (HR: 0.75, 95% CI 0.66 to 0.85) with CT, but statistical significance was not attained for the 7.5 mg dose (HR: 0.95, 95% CI 0.83 to 1.08). A significantly increased in PFS with doses of 5 mg (HR: 0.69, 95% CI 0.58 to 0.83), 7.5 mg (HR: 0.81, 95% CI 0.66 to 1.00), and 10 mg (HR: 0.60, 95% CI 0.53 to 0.68). ORR distinctly increased after 5 mg (RR: 1.34, 95% CI 1.15 to 1.55), 7.5 mg (RR: 1.25, 95% CI 1.05 to 1.50), and10 mg (RR: 2.27, 95% CI 1.82 to 2.84) doses were administered. Grade ≥ 3 AEs increased clearly in 5 mg (RR: 1.11, 95% CI 1.04 to 1.20) compared to 7.5 mg (RR: 1.05, 95% CI 0.82 to 1.35) and 10 mg (RR: 1.15, 95% CI 0.98 to 1.36). Bayesian analysis demonstrated that 10 mg Bev obtained the maximum time of OS (HR: 0.75, 95% CrI 0.58 to 0.97; probability rank = 0.05) indirectly compared to 5 mg and 7.5 mg Bev. Compared with 5 mg and 7.5 mg Bev, 10 mg Bev also holds the longest duration for PFS (HR: 0.59, 95% CrI 0.43 to 0.82; probability rank = 0.00). In terms of ORR, 10 mg Bev holds the maximum frequency (RR: 2.02, 95% CrI 1.52 to 2.66; probability rank = 0.98) in comparison to 5 mg and 7.5 mg Bev clearly. For grade ≥ 3 AEs, 10 mg Bev has the maximum incidence (RR: 1.15, 95% CrI 0.95 to 1.40, probability rank = 0.67) in comparison to other doses of Bev. CONCLUSION The study suggests that 10 mg dose Bev could be more effective in treating advanced CRC in efficacy, but 5 mg Bev could be more safer in terms of safety.
Collapse
Affiliation(s)
- Jia Deng
- College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xinglin Zeng
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenting Hu
- College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Tinghui Yue
- College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zicheng Luo
- College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Lian Zeng
- College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ping Li
- Colorectal and Anal Surgery, the First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No 71 Baoshan North Road, Guiyang, 550001, China.
| | - Jiang Chen
- Colorectal and Anal Surgery, the First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No 71 Baoshan North Road, Guiyang, 550001, China.
| |
Collapse
|
29
|
Knapen DG, de Haan JJ, Fehrmann RSN, de Vries EGE, de Groot DJA. Opportunities on the horizon for the management of early colon cancer. Crit Rev Oncol Hematol 2023; 183:103918. [PMID: 36702421 DOI: 10.1016/j.critrevonc.2023.103918] [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: 10/11/2022] [Revised: 12/05/2022] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
There is a clear unmet need to improve early colon cancer management. This review encompasses the current systemic treatment landscape and summarises novel and pivotal trials. The Immunoscore and circulating tumour DNA (ctDNA) are studied to evaluate which patients should receive no, 3, or 6 months of adjuvant treatment. Several trials also test escalating treatment strategies for non-cleared ctDNA following standard adjuvant chemotherapy. Advances made in treating patients with metastatic colon cancer are now being translated to the early colon cancer setting. Two ongoing RCTs study immune checkpoint inhibitors (ICI) in patients with microsatellite instable high (MSI-H) early colon cancer as adjuvant treatment. Neo-adjuvant treatment is being studied in several ongoing RCTs as well. The complete response rate in patients with MSI-H tumours following ICI in neoadjuvant trials has potential organ-sparing implications.
Collapse
Affiliation(s)
- Daan G Knapen
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Jacco J de Haan
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Derk Jan A de Groot
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
30
|
Tau S, Miller TW. The role of cancer cell bioenergetics in dormancy and drug resistance. Cancer Metastasis Rev 2023; 42:87-98. [PMID: 36696004 PMCID: PMC10233409 DOI: 10.1007/s10555-023-10081-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 01/11/2023] [Indexed: 01/26/2023]
Abstract
While anti-cancer drug treatments are often effective for the clinical management of cancer, these treatments frequently leave behind drug-tolerant persister cancer cells that can ultimately give rise to recurrent disease. Such persistent cancer cells can lie dormant for extended periods of time, going undetected by conventional clinical means. Understanding the mechanisms that such dormant cancer cells use to survive, and the mechanisms that drive emergence from dormancy, is critical to the development of improved therapeutic strategies to prevent and manage disease recurrence. Cancer cells often exhibit metabolic alterations compared to their non-transformed counterparts. An emerging body of evidence supports the notion that dormant cancer cells also have unique metabolic adaptations that may offer therapeutically targetable vulnerabilities. Herein, we review mechanisms through which cancer cells metabolically adapt to persist during drug treatments and develop drug resistance. We also highlight emerging therapeutic strategies to target dormant cancer cells via their metabolic features.
Collapse
Affiliation(s)
- Steven Tau
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Dartmouth Cancer Center, Lebanon, NH, USA
| | - Todd W Miller
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Dartmouth Cancer Center, Lebanon, NH, USA.
- Dartmouth-Hitchcock Medical Center, One Medical Center Drive, HB-7936, Lebanon, NH 03756, USA.
| |
Collapse
|
31
|
Boutros CS, Hue JJ, Elshami M, Rothermel LD, Hoehn RS, Ammori JB, Winter JM, Ocuin LM, Hardacre JM. Management of adenocarcinoma of the pancreatic tail in the elderly. J Surg Oncol 2023; 127:405-412. [PMID: 36301227 DOI: 10.1002/jso.27134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Elderly patients with adenocarcinoma of the pancreatic head can achieve reasonable survival with multimodal therapy. An analysis specific to cancers of the pancreatic tail has not been published. METHODS We identified patients ≥65 years with localized adenocarcinoma of the pancreatic tail in the National Cancer Database (2011-2017). Patients were grouped by age (65-79 and ≥80 years) and categorized by treatment regimen. Postoperative outcomes and survival were analyzed using propensity score matching and multivariable logistical regression. RESULTS 2168 patients were included: 73.9% were 65-79 years and 26.1% were ≥80 years. 34.1% of octogenarians did not receive any treatment, relative to 15.9% of younger patients (p < 0.001). Thirty-day mortality rates were similar in operatively managed patients; however, the 90-day mortality rate among octogenarians was greater (3.0% vs. 7.8%, p < 0.001; odds ratio [OR] = 1.85, 95% confidence interval [CI] = 1.07-3.19). Age ≥ 80 was not associated with survival on multivariable hazards regression (hazard ratio [HR] = 1.08, 95% CI = 0.95-1.24). After propensity matching, the addition of chemotherapy was not associated with improved survival relative to distal pancreatectomy alone among octogenarians (HR = 1.09, 95% CI = 0.72-1.65). CONCLUSIONS Management of adenocarcinoma of the pancreatic tail varies based on patient age. Resection appears to play a key role in management, but there is substantial upfront risk. Shared decision making should be employed to balance the chance for long-term survival with the risk of early mortality.
Collapse
Affiliation(s)
- Christina S Boutros
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Jonathan J Hue
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Mohamedraed Elshami
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Luke D Rothermel
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Richard S Hoehn
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - John B Ammori
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Jordan M Winter
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Lee M Ocuin
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Jeffrey M Hardacre
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
32
|
Collette F, Lawson G, Hassid S, Delahaut G, Bachy V, Van Der Vorst S, Faugeras L, Gilliaux Q, D'Hondt L. Aggressive recurrent respiratory papillomatosis: A series of five consecutive patients successfully treated with adjuvant intravenous bevacizumab. A single Belgian academic center experience. Head Neck 2023; 45:1071-1079. [PMID: 36840929 DOI: 10.1002/hed.27300] [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: 07/21/2022] [Revised: 12/22/2022] [Accepted: 01/23/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Recurrent respiratory papillomatosis (RRP) is a currently incurable benign neoplasm caused by human papilloma virus (HPV) infection. It usually reduces voice, respiratory, and general quality of life, and is sometimes life-threatening. Patients usually need repeated operations. The use of adjuvant bevacizumab, a monoclonal antibody targeting vascular endothelial growth factor A, has been described in several case reports, with a good efficacy and safety profile. METHODS We report the cases of five patients with aggressive RRP who were treated with adjuvant systemic bevacizumab in a single Belgian tertiary center. RESULTS A complete response was achieved in four patients after a median of 4.5 months, and a partial response in one. In all cases, the number of surgeries was drastically reduced, and quality of life improved. Toxicity was easily managed. CONCLUSIONS Systemic bevacizumab seems to be an effective and safe adjuvant treatment for aggressive RRP.
Collapse
Affiliation(s)
- Fanny Collette
- Department of Oncology, Université Catholique de Louvain, CHU UCL Namur, Yvoir, Belgium
| | - Georges Lawson
- Department of ENT and Head and Neck Surgery, Université Catholique de Louvain, CHU UCL Namur, Yvoir, Belgium
| | - Samantha Hassid
- Department of ENT and Head and Neck Surgery, Université Catholique de Louvain, CHU UCL Namur, Yvoir, Belgium
| | - Gilles Delahaut
- Department of ENT and Head and Neck Surgery, Université Catholique de Louvain, CHU UCL Namur, Yvoir, Belgium
| | - Vincent Bachy
- Department of ENT and Head and Neck Surgery, Université Catholique de Louvain, CHU UCL Namur, Yvoir, Belgium
| | - Sébastien Van Der Vorst
- Department of ENT and Head and Neck Surgery, Université Catholique de Louvain, CHU UCL Namur, Yvoir, Belgium
| | - Laurence Faugeras
- Department of Oncology, Université Catholique de Louvain, CHU UCL Namur, Yvoir, Belgium
| | - Quentin Gilliaux
- Department of Oncology, Université Catholique de Louvain, CHU UCL Namur, Yvoir, Belgium
| | - Lionel D'Hondt
- Department of Oncology, Université Catholique de Louvain, CHU UCL Namur, Yvoir, Belgium
| |
Collapse
|
33
|
Younginger BS, Mayba O, Reeder J, Nagarkar DR, Modrusan Z, Albert ML, Byrd AL. Enrichment of oral-derived bacteria in inflamed colorectal tumors and distinct associations of Fusobacterium in the mesenchymal subtype. Cell Rep Med 2023; 4:100920. [PMID: 36706753 PMCID: PMC9975273 DOI: 10.1016/j.xcrm.2023.100920] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 11/22/2022] [Accepted: 01/06/2023] [Indexed: 01/27/2023]
Abstract
While the association between colorectal cancer (CRC) features and Fusobacterium has been extensively studied, less is known of other intratumoral bacteria. Here, we leverage whole transcriptomes from 807 CRC samples to dually characterize tumor gene expression and 74 intratumoral bacteria. Seventeen of these species, including 4 Fusobacterium spp., are classified as orally derived and are enriched among right-sided, microsatellite instability-high (MSI-H), and BRAF-mutant tumors. Across consensus molecular subtypes (CMSs), integration of Fusobacterium animalis (Fa) presence and tumor expression reveals that Fa has the most significant associations in mesenchymal CMS4 tumors despite a lower prevalence than in immune CMS1. Within CMS4, the prevalence of Fa is uniquely associated with collagen- and immune-related pathways. Additional Fa pangenome analysis reveals that stress response genes and the adhesion FadA are commonly expressed intratumorally. Overall, this study identifies oral-derived bacteria as enriched in inflamed tumors, and the associations of bacteria and tumor expression are context and species specific.
Collapse
Affiliation(s)
- Brett S Younginger
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Oleg Mayba
- Department of OMNI Bioinformatics, Genentech, Inc., South San Francisco, CA, USA
| | - Jens Reeder
- Department of Oncology Bioinformatics, Genentech, Inc., South San Francisco, CA, USA
| | - Deepti R Nagarkar
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Zora Modrusan
- Microchemistry, Proteomics, Lipidomics and Next Generation Sequencing, Genentech, Inc., South San Francisco, CA, USA
| | | | - Allyson L Byrd
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
34
|
Zhu Y, Zhou X, Yao Z. A mini-review: Advances in plant-derived extracellular vesicles as nano-delivery systems for tumour therapy. Front Bioeng Biotechnol 2022; 10:1076348. [PMID: 36588940 PMCID: PMC9797590 DOI: 10.3389/fbioe.2022.1076348] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Extracellular vesicles are functionally active, nanoscale, membrane-bound vesicles that can be secreted by all cells. They have a key role in most health and disease states and have gradually become a promising class of delivery vehicles for targeted therapies for a variety of diseases. Plant-derived extracellular vesicles have received increasing attention based on their easy availability, non-toxicity and high absorption. However, compared with mammalian extracellular vesicles, the role of these nanoparticles as nano-delivery systems in tumour therapy has been underestimated. In this paper, the application of plant-derived extracellular vesicles and their nano-derivatives as nano-delivery systems in tumour therapy is reviewed to illustrate their great application potential.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Basic Medical, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiaona Zhou
- Department of First Clinical Medical, Yunnan University of Chinese Medicine, Kunming, China,*Correspondence: Zheng Yao, ; Xiaona Zhou,
| | - Zheng Yao
- Department of Basic Medical, Yunnan University of Chinese Medicine, Kunming, China,Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Kunming, China,*Correspondence: Zheng Yao, ; Xiaona Zhou,
| |
Collapse
|
35
|
Zheng H, McCombs JS, Barzi A. Provider imaging practices and outcomes of advanced colorectal cancer. Cancer Treat Res Commun 2022; 32:100624. [PMID: 36027699 DOI: 10.1016/j.ctarc.2022.100624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
PURPOSE To explore the real-world utilization of computerized tomography (CT) in patients with advanced colorectal cancer (CRC) and the associated outcomes. METHODS Using Optum's de-identified Clinformatics® Data Mart Database (2008-2016), we identified patients with CRC receiving combination of chemotherapies (fluoropyrimidines with either oxaliplatin or irinotecan, or capecitabine with either oxaliplatin or irinotecan) combined with bevacizumab as the initial treatment, and its starting date was registered as the index date. End of treatment was defined by the presence of a gap in therapy > 60 days or treatment switch. We assessed the CT scan utilization during the period between 60 days pre-index and 30 days post-end of treatment. Cox regressions were performed to assess the impact of intensity of follow-up CT scans, measured by time to the first scan from the index date, on likelihood of treatment switch and survival. RESULTS Among included 4,810 patients, the median (standard deviation) time to the first follow-up scan was 57 (45) days. The mean and median number of CT scans was 4 and 3, respectively. An earlier follow-up scan was associated with significantly greater chance of treatment switch (hazard ratio = 0.99 for each day of delay, P < 0.01), and greater likelihood of death (hazard ratio = 0.99 for each day of delay, P < 0.01). CONCLUSION More intense follow-up increased the likelihood of treatment switch, yet it was not associated with better survival outcome. Further analysis in populations with longer follow-up period is warranted to elucidate the link between CT scan utilization and outcomes.
Collapse
Affiliation(s)
- Hanke Zheng
- Department of Pharmaceutical and Health Economics, School of Pharmacy, University of Southern California, Los Angeles, CA, United States of America; Leonard D. Schaeffer Center for Health Policy and Economics, University of Southern California, Los Angeles, CA, United States of America
| | - Jeffrey S McCombs
- Department of Pharmaceutical and Health Economics, School of Pharmacy, University of Southern California, Los Angeles, CA, United States of America; Leonard D. Schaeffer Center for Health Policy and Economics, University of Southern California, Los Angeles, CA, United States of America
| | - Afsaneh Barzi
- Leonard D. Schaeffer Center for Health Policy and Economics, University of Southern California, Los Angeles, CA, United States of America; Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA, United States of America.
| |
Collapse
|
36
|
Riesco-Martinez MC, Modrego A, Espinosa-Olarte P, La Salvia A, Garcia-Carbonero R. Perioperative Chemotherapy for Liver Metastasis of Colorectal Cancer: Lessons Learned and Future Perspectives. Curr Treat Options Oncol 2022; 23:1320-1337. [PMID: 35980520 DOI: 10.1007/s11864-022-01008-5] [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] [Accepted: 08/01/2022] [Indexed: 11/27/2022]
Abstract
OPINION STATEMENT Colorectal cancer (CRC) is a major public health problem and the 2nd leading-cause of cancer-related death worldwide. Around 30% of patients present with metastatic disease and 50% of those with early disease will eventually relapse. The metastatic spread occurs mainly to the liver, which is the exclusive site in 30-40% of the cases. Surgery is the main curative option for liver recurrence, but only one out of five patients are eligible for resection. Moreover, even if surgery is feasible, recurrence rate is high, occurring in up to 75% of patients. Therefore, additional treatment to improve these disappointing outcomes has been sought. Adjuvant and perioperative chemotherapy aim to eradicate early micrometastatic disease, decreasing recurrence rates, and improving survival outcomes. Different chemotherapy regimens, mainly extrapolated from the adjuvant experience, have showed conflicting results, with improvements in disease free but not in overall survival. The addition of targeted therapies to chemotherapy has improved response rates and resectability when administered preoperatively, but did not have an impact on survival in the adjuvant setting. There is a need to critically synthetize the available evidence on perioperative and conversion therapy from the past years, and appraise areas of current research and potential future directions.
Collapse
Affiliation(s)
- Maria C Riesco-Martinez
- Medical Oncology Department, Hospital Universitario 12 de Octubre, imas 12, UCM, Avda Cordoba km 5.4, 28041, Madrid, Spain
| | - Andrea Modrego
- Medical Oncology Department, Hospital Universitario 12 de Octubre, imas 12, UCM, Avda Cordoba km 5.4, 28041, Madrid, Spain
| | - Paula Espinosa-Olarte
- Medical Oncology Department, Hospital Universitario 12 de Octubre, imas 12, UCM, Avda Cordoba km 5.4, 28041, Madrid, Spain
| | - Anna La Salvia
- Medical Oncology Department, Hospital Universitario 12 de Octubre, imas 12, UCM, Avda Cordoba km 5.4, 28041, Madrid, Spain
| | - Rocio Garcia-Carbonero
- Medical Oncology Department, Hospital Universitario 12 de Octubre, imas 12, UCM, Avda Cordoba km 5.4, 28041, Madrid, Spain.
| |
Collapse
|
37
|
Luo J, Zhong X, Peng Y, Hao C, Liang X, Yang Y, Shi X, Chen X, Yi X, Li X, Wu J, Li J, Xiao Q, Wu C, Lu R, Pan Y, Wang X, Fan JB, Wang Y, Wang Y. Self-anti-angiogenesis nanoparticles enhance anti-metastatic-tumor efficacy of chemotherapeutics. Bioact Mater 2022; 13:179-190. [PMID: 35224300 PMCID: PMC8843953 DOI: 10.1016/j.bioactmat.2021.10.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 12/22/2022] Open
Abstract
Beyond traditional endothelium-dependent vessel (EDV), vascular mimicry (VM) is another critical tumor angiogenesis that further forms in many malignant metastatic tumors. However, the existing anti-angiogenesis combined chemotherapeutics strategies are only efficient for the treatment of EDV-based subcutaneous tumors, but remain a great challenge for the treatment of in situ malignant metastatic tumor associated with EDV and VM. Here, we demonstrate a self-assembled nanoparticle (VE-DDP-Pro) featuring self-anti-EDV and -VM capacity enables to significantly enhance the treatment efficacy of cisplatin (DDP) against the growth and metastasis of ovarian cancer. The VE-DDP-Pro is constructed by patching DDP loaded cRGD-folate-heparin nanoparticles (VE) onto the surface of protamine (Pro) nanoparticle. We demonstrated the self-anti-angiogenesis capacity of VE-DDP-Pro was attributed to VE, which could significantly inhibit the formation of EDV and VM by regulating signaling pathway of MMP-2/VEGF, AKT/mTOR/MMP-2/Laminin and AKT/mTOR/EMT, facilitating chemotherapeutics to effectively suppress the development and metastasis of ovarian cancer. Thus, combing with the chemotherapeutics effectiveness of DDP, the VE-DDP-Pro can significantly enhance treatment efficacy and prolong median survival of mice with metastatic ovarian cancer. We believe our self-assembled nanoparticles integrating the anti-EDV and anti-VM capacity provide a new preclinical sight to enhance the efficacy of chemotherapeutics for the treatment malignant metastasis tumor.
Collapse
Affiliation(s)
- Jiamao Luo
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xinxian Zhong
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yingming Peng
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Chenyuan Hao
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiaomei Liang
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yulu Yang
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiubo Shi
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xuncai Chen
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xiao Yi
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaoxuan Li
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jianhua Wu
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jinheng Li
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qian Xiao
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Chentian Wu
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ruojing Lu
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yao Pan
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xuejiao Wang
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jun-Bing Fan
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yifeng Wang
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Ying Wang
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
38
|
Diasio RB, Offer SM. Testing for Dihydropyrimidine Dehydrogenase Deficiency to Individualize 5-Fluorouracil Therapy. Cancers (Basel) 2022; 14:3207. [PMID: 35804978 PMCID: PMC9264755 DOI: 10.3390/cancers14133207] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 12/24/2022] Open
Abstract
Severe adverse events (toxicity) related to the use of the commonly used chemotherapeutic drug 5-fluorouracil (5-FU) affect one in three patients and are the primary reason cited for premature discontinuation of therapy. Deficiency of the 5-FU catabolic enzyme dihydropyrimidine dehydrogenase (DPD, encoded by DPYD) has been recognized for the past 3 decades as a pharmacogenetic syndrome associated with high risk of 5-FU toxicity. An appreciable fraction of patients with DPD deficiency that receive 5-FU-based chemotherapy die as a result of toxicity. In this manuscript, we review recent progress in identifying actionable markers of DPD deficiency and the current status of integrating those markers into the clinical decision-making process. The limitations of currently available tests, as well as the regulatory status of pre-therapeutic DPYD testing, are also discussed.
Collapse
Affiliation(s)
- Robert B. Diasio
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA;
- Mayo Clinic College of Medicine and Science, Mayo Clinic, Rochester, MN 55902, USA
| | - Steven M. Offer
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA;
- Mayo Clinic College of Medicine and Science, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
39
|
Notch signaling regulates vessel structure and function via Hspg2. Gene 2022; 826:146439. [PMID: 35339643 DOI: 10.1016/j.gene.2022.146439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 03/18/2022] [Indexed: 11/20/2022]
Abstract
The abnormal structure of tumor blood vessels is an important reason for the low efficacy of anti-tumor drugs. Notch signaling is an evolutionarily highly conserved signaling pathway that plays an important role in vessel development. However, the role and mechanism of Notch signaling in the formation of vascular structure is not fully understood. In this study, we demonstrated that blocking Notch signaling in endothelial cells (ECs) leads to obstructed tumor blood vessel basement membrane formation and the reduction of blood perfusion, as well as blood-retinal barrier (BRB) and blood-brain barrier (BBB) destruction in healthy mice. Endothelial Notch overactivation exacerbates the increases in tumor blood vessel basement membrane and blood perfusion ratio, and promotes recruitment of retinal vascular smooth muscle cells in neonatal mice. Notch signaling also regulates the formation of adhesion junctions (AJs) in ECs. In addition, we confirmed that Notch signaling regulates the AJs of ECs by regulating the expression of downstream gene Hspg2. This research is of great theoretical and practical significance for understanding the mechanism of tumor vascular structure formation as well as the search for new targets for vascular-targeted therapy.
Collapse
|
40
|
New Insights into Adjuvant Therapy for Localized Colon Cancer. Hematol Oncol Clin North Am 2022; 36:507-520. [DOI: 10.1016/j.hoc.2022.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
41
|
Production and optimization of a vasostatin-30 and vasoinhibin fusion protein that inhibits tumor angiogenesis and dissemination of breast cancer cells in a zebrafish model. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
42
|
Patil V, Noronha V, Joshi A, Menon N, Mathrudev V, Bhattacharjee A, Chandrasekharan A, Vallathol D, Dsouza H, Srinivas S, Mandal T, Chaturvedi P, Chaukar D, Pai P, Nair S, Thiagrajan S, Laskar S, Nawale K, Babanrao Dhumal S, Tambe R, Banavali S, Prabhash K. RMAC study: A randomized study for evaluation of metronomic adjuvant chemotherapy in recurrent head and neck cancers post salvage surgical resection in those who are ineligible for re-irradiation. Oral Oncol 2022; 128:105816. [DOI: 10.1016/j.oraloncology.2022.105816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 10/18/2022]
|
43
|
A retrospective analysis using deep-learning models for prediction of survival outcome and benefit of adjuvant chemotherapy in stage II/III colorectal cancer. J Cancer Res Clin Oncol 2022; 148:1955-1963. [PMID: 35332389 DOI: 10.1007/s00432-022-03976-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Most of Stage II/III colorectal cancer (CRC) patients can be cured by surgery alone, and only certain CRC patients benefit from adjuvant chemotherapy. Risk stratification based on deep-learning from haematoxylin and eosin (H&E) images has been postulated as a potential predictive biomarker for benefit from adjuvant chemotherapy. However, very limited success has been achieved in using biomarkers, including deep-learning-based markers, to facilitate the decision for adjuvant chemotherapy despite recent advances of artificial intelligence. METHODS We trained and internally validated CRCNet using 780 Stage II/III CRC patients from Molecular and Cellular Oncology. Independent external validation of the model was performed using 337 Stage II/III CRC patients from The Cancer Genome Atlas (TCGA). RESULTS CRCNet stratified the patients into high, medium, and low-risk subgroups. Multivariate Cox regression analyses confirmed that CRCNet risk groups are statistically significant after adjusting for existing risk factors. The high-risk subgroup significantly benefits from adjuvant chemotherapy. A hazard ratio (chemo-treated vs untreated) of 0.2 (95% Confidence Interval (CI), 0.05-0.65; P = 0.009) and 0.6 (95% CI 0.42-0.98; P = 0.038) are observed in the TCGA and MCO Fluorouracil-treated patients, respectively. Conversely, no significant benefit from chemotherapy is observed in the low- and medium-risk groups (P = 0.2-1). CONCLUSION The retrospective analysis provides further evidence that H&E image-based biomarkers may potentially be of great use in delivering treatments following surgery for Stage II/III CRC, improving patient survival, and avoiding unnecessary treatment and associated toxicity, and warrants further validation on other datasets and prospective confirmation in clinical trials.
Collapse
|
44
|
Aoyama T, Yoshikawa T, Ida S, Cho H, Sakamaki K, Ito Y, Fujitani K, Takiguchi N, Kawashima Y, Nishikawa K, Nunobe S, Hiki N. Effects of perioperative eicosapentaenoic acid‑enriched oral nutritional supplement on the long‑term oncological outcomes after total gastrectomy for gastric cancer. Oncol Lett 2022; 23:151. [PMID: 35836480 PMCID: PMC9258592 DOI: 10.3892/ol.2022.13272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/15/2022] [Indexed: 12/24/2022] Open
Abstract
Basic and clinical reports have suggested that eicosapentaenoic acid (EPA) exhibits anti-tumor activity. The present study evaluated whether perioperative EPA could improve the survival of patients with localized gastric cancer as a key secondary endpoint of a randomized clinical study. The present study was designed as multicenter, open-label, superiority, randomized trial to confirm the preventive effect of EPA on body weight loss after total gastrectomy for gastric cancer. Eligible patients were randomized to either the standard-diet group (EPA-off group) or EPA-on group by a centralized dynamic method. An EPA-enriched supplement (ProSure®) was given to the EPA-on group in addition to their standard diet. This supplement included 600 kcal with 2.2 g/day of EPA. Among the 126 patients who were randomized, 123 patients (EPA-off group, n=60; EPA-on group, n=63) were examined in the survival analyses. All background factors were well balanced between the two groups. The 3-year and 5-year overall survival rates were 74.6 and 67.8%, respectively, in the EPA-off group, and 77.8 and 76.2% in the EPA-on group. There was no significant difference between the EPA-off and EPA-on groups (hazard ratio, 0.77; P=0.424). In the subgroup analysis, the hazard ratio was 0.39 in patients who received neoadjuvant chemotherapy and 0.57 in patients with nodal metastasis. In conclusion, a clear survival benefit of perioperative EPA was not observed in localized gastric cancer. The value of EPA should be further tested in a future study in patients with unfavorable advanced gastric cancer. Clinical trial number: UMIN000006380; date of registration, September 21, 2011.
Collapse
Affiliation(s)
- Toru Aoyama
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Kanagawa 241‑8515, Japan
| | - Takaki Yoshikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Kanagawa 241‑8515, Japan
| | - Satoshi Ida
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135‑0063, Japan
| | - Haruhiko Cho
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Kanagawa 241‑8515, Japan
| | - Kentaro Sakamaki
- Department of Biostatistics and Epidemiology, Yokohama City University Medical Center, Yokohama, Kanagawa 232‑0024, Japan
| | - Yuichi Ito
- Department of Gastroenterological Surgery, Aichi Cancer Center, Nagoya, Aichi 464‑8681, Japan
| | - Kazumasa Fujitani
- Department of Surgery, Osaka General Medical Center, Osaka 558‑8558, Japan
| | - Nobuhiro Takiguchi
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chuo-ku, Chiba 260‑8781, Japan
| | - Yoshiyuki Kawashima
- Department of Gastroenterological Surgery, Saitama Cancer Center, Kitaadachi, Saitama 362‑0806, Japan
| | | | - Soya Nunobe
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135‑0063, Japan
| | - Naoki Hiki
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135‑0063, Japan
| |
Collapse
|
45
|
|
46
|
Vogel JD, Felder SI, Bhama AR, Hawkins AT, Langenfeld SJ, Shaffer VO, Thorsen AJ, Weiser MR, Chang GJ, Lightner AL, Feingold DL, Paquette IM. The American Society of Colon and Rectal Surgeons Clinical Practice Guidelines for the Management of Colon Cancer. Dis Colon Rectum 2022; 65:148-177. [PMID: 34775402 DOI: 10.1097/dcr.0000000000002323] [Citation(s) in RCA: 133] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
| | | | | | | | | | | | - Amy J Thorsen
- Colon and Rectal Surgery Associates, Minneapolis, Minnesota
| | | | | | | | | | | |
Collapse
|
47
|
Inhibition of angiotensin pathway via valsartan reduces tumor growth in models of colorectal cancer. Toxicol Appl Pharmacol 2022; 440:115951. [PMID: 35235860 DOI: 10.1016/j.taap.2022.115951] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/09/2022] [Accepted: 02/23/2022] [Indexed: 01/05/2023]
|
48
|
Abstract
Most patients with colorectal cancer (CRC) were diagnosed in advanced stage and the prognosis is poor. Therefore, early detection and prevention of CRC are very important. As with other cancers, there is also the tertiary prevention for CRC. The primary prevention is etiological prevention, which is mainly the treatment of adenoma or inflammation for preventing the development into cancer. The secondary prevention is the early diagnosis and early treatment for avoiding progressing to advanced cancer. The tertiary prevention belongs to the broad category of prevention, mainly for advanced CRC, through surgical treatment and postoperative adjuvant chemotherapy, radiotherapy, targeted therapy, immunotherapy for preventing tumor recurrence or metastasis. This consensus is based on the recent domestic and international consensus guidelines and the latest progress of international researches in the past five years. This consensus opinion seminar was hosted by the Chinese Society of Gastroenterology and Cancer Collaboration Group of Chinese Society of Gastroenterology, and was organized by the Division of Gastroenterology and Hepatology & Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University. The consensus opinion contains 60 statement clauses, the standard and basis of the evidence-based medicine grade and voting grade of the statement strictly complied with the relevant international regulations and practice.
Collapse
|
49
|
Asada Y, Chinen K, Yamataka K, Tokuyama J, Kurihara N, Iida S. Pathological complete response after neoadjuvant chemotherapy with FOLFOX for locally advanced sigmoid colon cancer with diverticulitis: A case report. Int J Surg Case Rep 2022; 90:106685. [PMID: 34952318 PMCID: PMC8714994 DOI: 10.1016/j.ijscr.2021.106685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/07/2021] [Accepted: 12/14/2021] [Indexed: 11/28/2022] Open
Abstract
Introduction and importance The standard treatment for locally advanced colon cancer (LACC) without distant metastasis is curative surgery followed by adjuvant chemotherapy, but the long-term outcomes of this strategy are not satisfactory. Neoadjuvant chemotherapy (NAC) is a promising novel option to overcome this issue. Tumor regression is an expected effect of NAC for LACC, but pathological complete response (pCR) is rare. In this report, we present a rare case of pCR after NAC with FOLFOX for LACC in the sigmoid colon. Presentation of case A 66-year-old woman presented to our hospital with fever and abdominal pain. The diagnosis was LACC in the sigmoid colon with possible invasion of the uterus and pelvic wall, stage IIIC (T4bN1bM0). Furthermore, the tumor was complicated by diverticulitis. A colostomy was performed, followed by NAC with FOLFOX. Six cycles were completed without significant adverse events, and the lesion shrunk remarkably. We performed a curative sigmoidectomy without any postoperative complications. Pathological examination revealed no viable cancer cells, indicating pCR. Discussion To the best of our knowledge, this is the first report of pCR after NAC for LACC complicated by diverticulitis. Colostomy before NAC, regimen, and cycle of NAC may be the key to this favorable course. Conclusion We present a rare case of pathological complete after neoadjuvant chemotherapy with FOLFOX for locally advanced colon cancer in the sigmoid colon complicated by diverticulitis. Our experience may be valuable in determining the optimal treatment strategy for LACC complicated by diverticulitis. Neoadjuvant chemotherapy is a promising novel option for locally advanced colon cancer. Pathological complete response is rare. Indications for cases complicated with diverticulitis are unknown. We present a rare case of pCR after NAC for LACC complicated by diverticulitis.
Collapse
Affiliation(s)
- Yusuke Asada
- Department of Surgery, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan.
| | - Katsuya Chinen
- Department of Pathology, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan
| | - Ken Yamataka
- Department of Surgery, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan
| | - Jo Tokuyama
- Department of Surgery, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan
| | - Naoto Kurihara
- Department of Surgery, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan
| | - Shuhei Iida
- Department of Surgery, Nerima General Hospital, 1-24-1 Asahigaoka, Nerima, Tokyo 176-8530, Japan
| |
Collapse
|
50
|
Transcriptomic profiling of adjuvant colorectal cancer identifies three key prognostic biological processes and a disease specific role for granzyme B. PLoS One 2022; 16:e0262198. [PMID: 34972191 PMCID: PMC8719661 DOI: 10.1371/journal.pone.0262198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 11/25/2021] [Indexed: 12/20/2022] Open
Abstract
Background Colorectal cancer (CRC) is a leading cause of cancer-related deaths, with a 5% 5-year survival rate for metastatic disease, yet with limited therapeutic advancements due to insufficient understanding of and inability to accurately capture high-risk CRC patients who are most likely to recur. We aimed to improve high-risk classification by identifying biological pathways associated with outcome in adjuvant stage II/III CRC. Methods and findings We included 1062 patients with stage III or high-risk stage II colon carcinoma from the prospective three-arm randomized phase 3 AVANT trial, and performed expression profiling to identify a prognostic signature. Data from validation cohort GSE39582, The Cancer Genome Atlas, and cell lines were used to further validate the prognostic biology. Our retrospective analysis of the adjuvant AVANT trial uncovered a prognostic signature capturing three biological functions—stromal, proliferative and immune—that outperformed the Consensus Molecular Subtypes (CMS) and recurrence prediction signatures like Oncotype Dx in an independent cohort. Importantly, within the immune component, high granzyme B (GZMB) expression had a significant prognostic impact while other individual T-effector genes were less or not prognostic. In addition, we found GZMB to be endogenously expressed in CMS2 tumor cells and to be prognostic in a T cell independent fashion. A limitation of our study is that these results, although robust and derived from a large dataset, still need to be clinically validated in a prospective study. Conclusions This work furthers our understanding of the underlying biology that propagates stage II/III CRC disease progression and provides scientific rationale for future high-risk stratification and targeted treatment evaluation in biomarker defined subpopulations of resectable high-risk CRC. Our results also shed light on an alternative GZMB source with context-specific implications on the disease’s unique biology.
Collapse
|