1
|
Nakano Y, Nishikawa G, Degawa K, Moriyoshi K, Kuriyama K, Watanabe Y, Miyamoto S. A case of multiple advanced colon cancers with spontaneous regression of only one lesion after biopsy: a case report and literature review. Clin J Gastroenterol 2025; 18:393-398. [PMID: 40029573 DOI: 10.1007/s12328-025-02106-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/09/2025] [Indexed: 03/05/2025]
Abstract
A 90-year-old man underwent a colonoscopy due to abdominal distension, revealing half-circumferential Type 2 advanced cancers in the ascending and transverse colon. No distant metastasis was detected, and 3 months later, laparoscopic extended right hemicolectomy was performed for both lesions. Pathological examination revealed ulcers and mucus retention in the ascending colon lesion without tumor components. A small number of signet ring cell-like tumor cells were found in the regional lymph node of the ascending colon, while well-to-moderately differentiated tubular adenocarcinoma was observed on the serosal surface in the transverse colon. Tumor regression was observed in the ascending colon cancer and lymph-node metastasis. Mismatch repair (MMR) protein immunostaining was conducted on biopsy tissues from both lesions. The ascending colon lesion showed weak positivity for MLH1, positivity for MSH2 and MSH6, and negativity for PMS2, indicating MMR deficiency, whereas the transverse colon lesion showed positivity for all of them, indicating MMR-proficient tumor. This is the first case report of multiple advanced colon cancers, where only one lesion exhibited spontaneous regression after biopsy, suggesting a potential link between MMR deficiency and the spontaneous regression of colorectal cancer.
Collapse
Affiliation(s)
- Yoshiko Nakano
- Department of Gastroenterology, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-Cho, Fukakusa, Fushimi-Ku, Kyoto, Japan
| | - Gen Nishikawa
- Department of Surgery, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-Cho, Fukakusa, Fushimi-Ku, Kyoto, Japan
| | - Kanako Degawa
- Department of Surgery, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-Cho, Fukakusa, Fushimi-Ku, Kyoto, Japan
| | - Koki Moriyoshi
- Department of Diagnostic Pathology, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-Cho, Fukakusa, Fushimi-Ku, Kyoto, Japan
| | - Katsutoshi Kuriyama
- Department of Gastroenterology, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-Cho, Fukakusa, Fushimi-Ku, Kyoto, Japan
| | - Yasuhiro Watanabe
- Department of Gastroenterology, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-Cho, Fukakusa, Fushimi-Ku, Kyoto, Japan
| | - Shin'ichi Miyamoto
- Department of Gastroenterology, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-Cho, Fukakusa, Fushimi-Ku, Kyoto, Japan.
| |
Collapse
|
2
|
Güven DC, Thong MS, Arndt V. Survivorship outcomes in patients treated with immune checkpoint inhibitors: a scoping review. J Cancer Surviv 2025; 19:806-845. [PMID: 38175366 PMCID: PMC12081552 DOI: 10.1007/s11764-023-01507-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have become a central part of cancer care. However, the survivorship outcomes in patients treated with ICIs are understudied. Therefore, we conducted a scoping review to evaluate the current status of the field and to establish research gaps regarding survivorship outcomes with ICIs in real-life cohorts. METHODS We used the Web of Science, PubMed, and Embase databases to systematically filter published studies with real-life cohorts from January 1, 2010, until October 19, 2022. Studies evaluating at least one survivorship outcome in ICI-treated patients were included. RESULTS A total of 39 papers were included. Quality of life (QoL) (n = 23), toxicity burden (n = 16), and psychosocial issues (n = 9) were the most frequently evaluated survivorship outcomes. Anti-PD-1/PD-L1 monotherapy and a response to treatment were associated with better QoL. In addition, the ICIs were associated with grade 3 or higher immune-related adverse events (irAEs) in 10-15% and late/long-term irAEs in 20-30% of the survivors. Regarding psychosocial problems, over 30% of survivors showed evidence of anxiety and depression, and 30-40% of survivors reported neurocognitive impairments. CONCLUSION The survivors treated with ICIs have impairments in most survivorship domains. Further research is needed to gather data on the understudied survivorship outcomes like late and long-term effects, fertility, financial toxicity, and return to work in survivors treated with ICIs. IMPLICATIONS FOR CANCER SURVIVORS Available evidence demonstrates that a significant portion of survivors treated with ICIs have a significant toxicity burden, lower QoL than the general population, and a high rate of psychosocial problems.
Collapse
Affiliation(s)
- Deniz Can Güven
- Department of Medical Oncology, Hacettepe University Cancer Institute, 06100 Sihhiye, Ankara, Turkey.
- Health Sciences University, Elazig City Hospital, Elazig, Turkey.
- Unit of Cancer Survivorship, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Melissa Sy Thong
- Unit of Cancer Survivorship, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Volker Arndt
- Unit of Cancer Survivorship, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
3
|
Flanagan M, Kelly M, McCormick P. Autoamputation in Mismatch Repair-Deficient Colon Cancer. JAMA Oncol 2025:2834391. [PMID: 40402493 DOI: 10.1001/jamaoncol.2025.1323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
A woman in her 50s with a history of hemochromatosis, psoriasis, and type 2 diabetes presented with a 3-month history of abdominal pain, weight loss, and a large palpable fixed abdominal mass. What is your diagnosis?
Collapse
Affiliation(s)
- Michael Flanagan
- Department of Colorectal Surgery, St James's Hospital, Dublin, Ireland
| | - Michael Kelly
- Department of Colorectal Surgery, St James's Hospital, Dublin, Ireland
| | - Paul McCormick
- Department of Colorectal Surgery, St James's Hospital, Dublin, Ireland
| |
Collapse
|
4
|
Cui HX, Yang XQ, Zhao GY, Wang FJ, Liu X. The neoadjuvant immunotherapy for non-metastatic mismatch repair-deficient colorectal cancer: a systematic review. Front Immunol 2025; 16:1540751. [PMID: 40376001 PMCID: PMC12078204 DOI: 10.3389/fimmu.2025.1540751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/07/2025] [Indexed: 05/18/2025] Open
Abstract
Background Immunotherapy has become the first-line treatment for metastatic mismatch repair deficient (dMMR) colorectal cancer. The efficacy and safety of neoadjuvant immunotherapy for the treatment of non-metastatic dMMR colorectal cancer remain unclear. In this article, we explore the clinical effect and safety of neoadjuvant immunotherapy for non-metastatic dMMR colorectal cancer. Methods We collected clinical data from the databases (PubMed, Wanfang Embase, Cochrane Library, and China National Knowledge Infrastructure databases) up to November 2024. The primary outcomes of major pathological response (MPR), pathological complete response (pCR), and other outcomes were analyzed for the final results. The secondary outcomes (pCR rates for the subgroups) were also analyzed. Results We included 21 articles with 628 non-metastatic dMMR colorectal cancers. A pCR was found in 320/480 (66.6%) patients [effect size (ES): 0.70, 95% CI: 0.66 to 0.74] with the fixed-effects model and little heterogeneity. A MPR was found in 388/452 (85.8%) patients (ES: 0.86, 95% CI: 0.81 to 0.91) with the fixed-effects model and little heterogeneity. In the subgroup analysis, pCR rates were similar in the T1-T3 group and T4a-T4b group in the fixed-effects model with minimal heterogeneity (OR: 0.76, 95% CI: 0.48 to 1.22). The pCR rates were similar in the colon cancer group and rectal cancer group in the fixed-effects model with minimal heterogeneity (OR: 1.41, 95% CI: 0.39 to 5.12). Similar pCR rates were found in the immune monotherapy group and immune therapy plus chemotherapy group (OR: 0.74, 95% CI: 0.26 to 2.10) with the fixed-effects model and little heterogeneity. Conclusion Neoadjuvant immunotherapy achieves high rates of pCR and MPR for non-metastatic dMMR colorectal cancer. For locally advanced T4 stage dMMR colorectal cancer, neoadjuvant immunotherapy can still achieve good pCR rates. Neoadjuvant immune monotherapy can achieve good pCRs rates, avoiding the toxic side effects caused by combined dual immunotherapy and chemo(radio)therapy. Neoadjuvant immunotherapy could be another treatment option for non-metastatic dMMR colorectal cancer. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024594173.
Collapse
Affiliation(s)
- Hong-Xia Cui
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Xiao-Quan Yang
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Guang-yue Zhao
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Feng-jian Wang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Xin Liu
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| |
Collapse
|
5
|
Cercek A, Foote MB, Rousseau B, Smith JJ, Shia J, Sinopoli J, Weiss J, Lumish M, Temple L, Patel M, Wilde C, Saltz LB, Argiles G, Stadler Z, Artz O, Maron S, Ku G, Gu P, Janjigian YY, Molena D, Iyer G, Coleman J, Abida W, Cohen S, Soares K, Schattner M, Strong VE, Yaeger R, Paty P, Shcherba M, Sugarman R, Romesser PB, Zervoudakis A, Desai A, Segal NH, El Dika I, Widmar M, Wei I, Pappou E, Fumo G, Aparo S, Gonen M, Gollub M, Jayaprakasham VS, Kim TH, Garcia Aguilar J, Weiser M, Diaz LA. Nonoperative Management of Mismatch Repair-Deficient Tumors. N Engl J Med 2025. [PMID: 40293177 DOI: 10.1056/nejmoa2404512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
BACKGROUND Among patients with mismatch repair-deficient (dMMR), locally advanced rectal cancer, neoadjuvant checkpoint blockade eliminated the need for surgery in a high proportion of patients. Whether this approach can be extended to all early-stage dMMR solid tumors, regardless of tumor site, is unknown. METHODS We conducted a phase 2 study in which patients with stage I, II, or III dMMR solid tumors that were amenable to curative-intent surgery were treated with neoadjuvant dostarlimab, a programmed cell death 1 (PD-1) blocking agent, for 6 months. The response to treatment was assessed in two cohorts: patients in cohort 1 had dMMR, locally advanced rectal cancer, and patients in cohort 2 had dMMR nonrectal solid tumors. Patients with a clinical complete response could elect to proceed with nonoperative management; those with residual disease were to undergo resection. In this analysis, the primary end point, assessed in cohort 1, was a sustained clinical complete response at 12 months. Recurrence-free survival and safety were evaluated. RESULTS A total of 117 patients were included in the analysis. In cohort 1, all 49 patients who completed treatment had a clinical complete response and elected to proceed with nonoperative management. A total of 37 patients had a sustained clinical complete response at 12 months, a finding that met the criterion for efficacy. In cohort 2, a total of 35 of 54 patients who completed treatment had a clinical complete response, and 33 elected to proceed with nonoperative management. Among the 103 patients who completed treatment across both cohorts, 84 had a clinical complete response, and 82 did not undergo surgery. Among the 117 total patients, recurrence-free survival at 2 years was 92% (95% confidence interval, 86 to 99); the median follow-up for recurrence was 20.0 months (range, 0 to 60.8). The majority of patients (95%) had reversible, grade 1 or 2 adverse events (60%) or had no adverse events (35%). The option for curative resection was not compromised during or after treatment in any of the patients. CONCLUSIONS Among patients with early-stage dMMR solid tumors that were amenable to curative-intent surgery, neoadjuvant PD-1 blockade led to organ preservation in a high proportion of patients. (Funded by Swim Across America and others; ClinicalTrials.gov number, NCT04165772.).
Collapse
Affiliation(s)
- Andrea Cercek
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Michael B Foote
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Benoit Rousseau
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - J Joshua Smith
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - Jenna Sinopoli
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Jill Weiss
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Melissa Lumish
- Department of Oncology, Case Comprehensive Cancer Center, Cleveland
| | - Lindsay Temple
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Miteshkumar Patel
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Callahan Wilde
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Leonard B Saltz
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Guillem Argiles
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Zsofia Stadler
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Oliver Artz
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Steven Maron
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Geoffrey Ku
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Ping Gu
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Yelena Y Janjigian
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Daniela Molena
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Gopa Iyer
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Jonathan Coleman
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Wassim Abida
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Seth Cohen
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Kevin Soares
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Mark Schattner
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Vivian E Strong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Rona Yaeger
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Philip Paty
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Marina Shcherba
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Ryan Sugarman
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Paul B Romesser
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Alice Zervoudakis
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Avni Desai
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Neil H Segal
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Imane El Dika
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Maria Widmar
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Iris Wei
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Emmanouil Pappou
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Gerard Fumo
- Department of Oncology, Hartford HealthCare, Hartford, CT
| | - Santiago Aparo
- Department of Oncology, Baptist Health Miami Cancer Institute, Miami
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Marc Gollub
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York
| | | | - Tae-Hyung Kim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York
| | | | - Martin Weiser
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Luis A Diaz
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| |
Collapse
|
6
|
Negoi I, Monson JRT, Bustamante-Lopez L, Garoufalia Z, D'Andrea V, Emile SH. Editorial: Organ preservation for rectal cancer patients. Front Surg 2025; 12:1600115. [PMID: 40352304 PMCID: PMC12061697 DOI: 10.3389/fsurg.2025.1600115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Affiliation(s)
- Ionut Negoi
- Department of General Surgery, Clinical Emergency Hospital of Bucharest, Carol Davila University of Medicine and Pharmacy Bucharest, Bucharest, Romania
| | - John R. T. Monson
- Department of Colorectal Surgery, Northshore University Hospital, Northwell Health, New York, NY, United States
| | - Leonardo Bustamante-Lopez
- Department of Colon & Rectum Surgery, AdventHealth, Orlando, FL, United States
- Surgical Health Outcomes Consortium (SHOC), Advent Health Digestive Institute, Orlando, FL, United States
| | - Zoe Garoufalia
- Colorectal Surgery Department, Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL, United States
| | - Vito D'Andrea
- Department of Surgery, Sapienza University of Rome, Rome, Italy
| | - Sameh Hany Emile
- Colorectal Surgery Department, Ellen Leifer Shulman and Steven Shulman Digestive Disease Center, Cleveland Clinic Florida, Weston, FL, United States
| |
Collapse
|
7
|
Lu Y, Chen L, Lin Y, Zhang Y, Wang Y, Yu W, Ren F, Guo H. Fatty acid metabolism: The crossroads in intestinal homeostasis and tumor. Metabolism 2025; 169:156273. [PMID: 40280478 DOI: 10.1016/j.metabol.2025.156273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/09/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Fatty acids (FAs) have various functions on cell regulation considering their abundant types and metabolic pathways. In addition, the relation between FA and other nutritional metabolism makes their functions more complex. As the first place for diet-derived FA metabolism, intestine is significantly influenced despite lack of clear conclusions due to the inconsistent findings. In this review, we discuss the regulation of fatty acid metabolism on the fate of intestinal stem cells in homeostasis and disorders, and also focus on the intestinal tumor development and treatment from the aspect of gut microbiota-epithelium-immune interaction. We summarize that the balances between FA oxidation and glycolysis, between oxidative phosphorylation and ketogenesis, between catabolism and anabolism, and the specific roles of individual FA types determine the diverse effects of intestinal FA metabolism in different cases. We hope this will inspire further dissection and suggest precise dietary/metabolic intervention for different demands related to intestinal health.
Collapse
Affiliation(s)
- Yao Lu
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Lining Chen
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yingying Lin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yafei Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yuqi Wang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Weiru Yu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Huiyuan Guo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
8
|
Feng J, Niu W, Zhang J, Ding Y, Li Z, Zhang J, Li B, Li C, Wang F, Wang G, Yu B. Pathological complete response after monotherapy with immune checkpoint inhibitors for bifocal colon cancer in a patient with lynch syndrome and situs inversus totalis: a case report. Front Immunol 2025; 16:1571607. [PMID: 40330463 PMCID: PMC12052791 DOI: 10.3389/fimmu.2025.1571607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Background Lynch syndrome is the most common hereditary colorectal cancer (CRC) syndrome, accounting for 3-5% of all CRC cases. Situs inversus totalis (SIT) is a rare congenital malformation with an incidence of 1 in 8,000 to 1 in 25,000. The co-occurrence of Lynch syndrome and SIT is extremely uncommon. Immune checkpoint inhibitors (ICIs) have demonstrated significant efficacy in treating microsatellite instability-high/deficient mismatch repair (MSI-H/dMMR) CRC. Tumors associated with Lynch syndrome frequently exhibit MSI-H, providing a theoretical basis for ICI use. Case presentation We report a case of bifocal colon cancer associated with Lynch syndrome and SIT. After seven cycles of sintilimab, the patient developed gastrointestinal perforation due to tumor regression, necessitating emergency surgery. The anatomical variations associated with SIT required the surgical team to adopt an alternative approach. Postoperatively, the patient continued sintilimab treatment for 2 years. In June 2024, he underwent a colostomy reversal and proximal colectomy. Pathological examination revealed a tumor regression grade (TRG) of 0, indicating complete pathological remission (pCR), with no recurrence or metastasis detected upon follow-up. Conclusions The anatomical variations associated with SIT increase the complexity of surgical procedures. Advanced imaging modalities such as computed tomography (CT) and magnetic resonance imaging (MRI) are essential for assessing fine anatomical details and facilitating surgery. ICIs are an effective treatment option for Lynch syndrome-associated CRC, as demonstrated in this case. Future studies should investigate the optimal timing of immunotherapy, combination treatment strategies, and methods to mitigate immune-related toxicities. Such research will help develop comprehensive and personalized treatment plans for Lynch syndrome-associated CRC.
Collapse
Affiliation(s)
- Jun Feng
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wenbo Niu
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Juan Zhang
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuanyi Ding
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zheng Li
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jianfeng Zhang
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Baokun Li
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chenhui Li
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Feifei Wang
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Guiying Wang
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Etiology Tracing and Individualized Diagnosis and Treatment for Digestive System Carcinoma, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Departments of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Bin Yu
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
9
|
Kang B, Qiao Y, Zhu J, Li J. Neoadjuvant therapy for colorectal cancer from 2015 to 2024: a visual analysis and bibliometric analysis. Front Oncol 2025; 15:1526610. [PMID: 40242245 PMCID: PMC11999843 DOI: 10.3389/fonc.2025.1526610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Background Colorectal cancer (CRC) imposes a substantial burden on global health., but research trends and hotspots in this field are still not clear. The purpose of this research is to create a visual knowledge map based on bibliometric analysis, identify research hotspots and predict future research trends. Method Utilizing the Web of Science Core Collection (WoSCC) as data source and integrating the visualization capabilities of the Bibliometrix R software package, CiteSpace, and VOSviewer, analyze the authors, research institutions, countries, cited documents, publishing journals, abstracts, and keyword information of literature pertaining to neoadjuvant therapy for colorectal cancer spanning from January 2015 to December 2024. Result The analysis included 1,587 articles from 1,464 institutions, 385 journals, and 61 countries or regions. China has the largest number of publications (449) and the largest number of citations (5,035). The United States occupies the leading position with an average of 21.6. "Annals of Surgical Oncology" is the most published journal with 51 articles, and "Journal of Clinical Oncology" is the journal with the most references (4,465 references). Highly cited references focus on clinical trials and guidelines for neoadjuvant therapy for colorectal cancer. In recent years, the most important keywords in the research on colorectal cancer and neoadjuvant therapy have been "artificial intelligence", "total neoadjuvant therapy" and "immunotherapy". Conclusion This article provided a review of the research on neoadjuvant therapy for colorectal cancer, can provide reference for subsequent research on neoadjuvant therapy for colorectal cancer. The results offered valuable insights and data that informed the direction of future advancements.
Collapse
Affiliation(s)
- Boyu Kang
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Yihuan Qiao
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Jun Zhu
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
- Department of General Surgery, The Southern Theater Air Force Hospital, Guangzhou, China
| | - Jipeng Li
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
- Department of Experiment Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
10
|
Palli E, Lavigne M, Verginis P, Alissafi T, Anastasopoulou A, Lyrarakis G, Kirkwood JM, Gogas H, Ziogas DC. Transcriptomic signatures in peripheral CD4 +T-lymphocytes may reflect melanoma staging and immunotherapy responsiveness prior to ICI initiation. Front Immunol 2025; 16:1529707. [PMID: 40226614 PMCID: PMC11986426 DOI: 10.3389/fimmu.2025.1529707] [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: 11/17/2024] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Background and purpose Promoting adaptive immunity with ICIs has drastically improved melanoma prognosis, but not for all patients. Some cases relapse in the first few months, while others keep durable benefit, even after immunotherapy discontinuation. To identify cellular/molecular signatures in peripheral blood that could differentiate advanced from metastatic melanoma and predict dynamics for primary/secondary immune escape, we examined 100 consecutive patients with stage III/IV melanoma scheduled to start ICIs. Materials and methods At melanoma diagnosis, a multiparameter flow cytometric analysis and purification scheme using standard conjugated antibodies were performed for all individuals prior to ICI initiation. In each stage(III/IV) according to their RFS/PFS, we retrospectively selected the cases with the clearest clinical outcomes and focused our analysis on the extreme responders(n=7) and non-responders(n=7) to characterize the transcriptomes of circulating CD4+T-cells by bulk RNA-seq, Differential Expression Analysis(DEA)and Gene Ontology(GO)enrichment analysis. Based on our selected patient cohort, we examined for differentially expressed genes(DEGs)and key-pathways that appear preferentially activated in stage III vs. IV melanoma, and in long vs. short immunotherapy responders. Results Although circulating immune-cells did not numerically differ in both sets of analysis(staging and ICI responsiveness), DEA and GO data showed that patients could be clustered separately, identifying 189vs.92 DEGs in stage IV/III and 101vs.47 DEGs in early progressors/long responders. These DEGs were functionally implicated in distinct pathways. For metastatic cases: inflammatory response(logp-value=-9.2:ADGRE5/2,CYBA,GRN,HMOX1,IRF5,ITGAM), adaptive immunity(logp-value=-7.7:CD1C,CD74,CYBB,NCF2,CTSA,S100A8/9,BCL3,FCER1G), T-cell activation(logp-value=-6.3:BCL3,CD1C,CD74,FCER1G,FGL2)and lipid metabolism/catabolism(logp-value=-2.5/-2.6:ARF3,GPX1,MVD,OCRL,PCCB,CTSA,PNPLA2,NAGLU,GBA2,ABHD4); while in early-progressors to ICIs: immune effector processing(logp-value=-13.7:BCL6,FGR,HLA-DQA1/DQB1,HLA-DRA,HLA-DRB1/DRB5,NKG7,SLC11A1,TYROBP,SPON2,HAVCR2),PD-1(logp-value=-10.2:HLA-DQA1/DQB1,HLA-DRA,HLA-DRB1/DRB5)and IFN signaling(logp-value=-8.5: HLA-DQA1/DQB1,HLA-DRA,HLA-DRB1/DRB5,NCAM1,IFITM3),positive regulation of T-cell activation(logp-value=-7.7:BCL6,HLA-DQA1/DQB1,HLA-DRA,HLA-DRB1/DRB5,SASH3,HAVCR2)and CD28 co-stimulation(logp-value=-10.3:HLA-DQA1/DQB1,HLA-DRA,HLA-DRB1/DRB5), supporting an immune-mediated behavior. Conclusions Specific pathways and marker genes in the peripheral CD4+T-cells may predetermine melanoma staging and immunotherapy resistance.
Collapse
Affiliation(s)
- Eleni Palli
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| | - Matthieu Lavigne
- Institute of Molecular Biology and Biotechnology of the Foundation for Research and Technology - Biology Department, University of Crete, School of Medicine, Heraklion, Greece
| | - Panagiotis Verginis
- Institute of Molecular Biology and Biotechnology of the Foundation for Research and Technology - Biology Department, University of Crete, School of Medicine, Heraklion, Greece
| | - Themis Alissafi
- Laboratory of Biology, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| | - Amalia Anastasopoulou
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| | - Georgios Lyrarakis
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| | - John M. Kirkwood
- Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, United States
| | - Helen Gogas
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| | - Dimitrios C. Ziogas
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens - School of Medicine, Athens, Greece
| |
Collapse
|
11
|
Duan J, Chen T, Li Q, Zhang Y, Lu T, Xue J, Sun Y, Gao L, Zhang Y. Protein arginine methyltransferase 6 enhances immune checkpoint blockade efficacy via the STING pathway in MMR-proficient colorectal cancer. J Immunother Cancer 2025; 13:e010639. [PMID: 40086819 PMCID: PMC11907083 DOI: 10.1136/jitc-2024-010639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The emergence of immunotherapy has revolutionized the paradigm of cancer treatment with immune checkpoint blockades (ICB) in solid cancers, including colorectal cancer (CRC). However, only a small subset of CRC patients harboring deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) benefits from ICB therapy. A very limited response to ICB therapy has been achieved in MMR-proficient CRC, representing a significant challenge limiting the clinical application of immunotherapy. MMR is the critical DNA repair pathway that maintains genomic integrity by correcting DNA mismatches, which is mediated by the MutSα or MutSβ complex consisting of MSH2 with MSH6 and MSH3, respectively. Given that MMR status directs effective immune response, we sought to determine whether targeting MMR capacity boosts ICB efficacy. METHODS Azoxymethane/dextran sodium sulfate (AOM/DSS)-induced CRC and xenograft model were used to evaluate the function of PRMT6 and response to PRMT6 inhibitor EPZ020411 and combination therapy of PD1 and EPZ020411. Biochemical assays were performed to elucidate the underlying mechanism of PRMT6-mediated MSH2 methylation and immune evasion. RESULTS We have identified PRMT6 as a crucial regulator of MMR capacity via MSH2 dimethylation at R171 and R219. Such a modification abrogates its MMR capacity and prevents the recruitment of MSH3 and MSH6. PRMT6 loss or inhibition triggers cytosolic DNA accumulation and cGAS-STING signaling activation, leading to enhanced immune response in PRMT6-deficient colon tumors or xenografts. Pharmacological inhibition of PRMT6 using EPZ020411 promotes mutagenesis and destabilizes MutSα or MutSβ assembly, and prolonged EPZ020411 exposure maintains an MSI-like phenotype in microsatellite stability (MSS) cells. EPZ020411 treatment sensitizes ICB efficacy of MSS cells, but not MSI cells in vivo. Similar effects have been observed in MSS colon tumors induced by AOM/DSS. CONCLUSIONS Our study provides a preclinical proof of concept to overcome resistance to immunotherapy by targeting PRMT6 in CRC with MSS.
Collapse
Affiliation(s)
- Jinlin Duan
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Pathology, Tongren Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tao Chen
- Department of Biliary-Pancreatic Surgery, Shanghai Jiao Tong University, Shanghai, China
| | - Qiwei Li
- Department of Biliary-Pancreatic Surgery, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Zhang
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital, Shanghai, China
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Ting Lu
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Junyan Xue
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yang Sun
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Ling Gao
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yonglong Zhang
- Laboratory of Targeted Therapy and Precision Medicine, Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
- Department of General Surgery, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
12
|
Cotte E, Arquilliere J, Artru P, Bachet JB, Benhaim L, Bibeau F, Christou N, Conroy T, Doyen J, Hoeffel C, Meillan N, Mirabel X, Pioche M, Rivin Del Campo E, Vendrely V, Huguet F, Bouché O. Rectal cancer - French intergroup clinical practice guidelines for diagnosis, treatment, and follow-up (TNCD, SNFGE, FFCD, GERCOR, UNICANCER, SFCD, SFED, SFRO, ACHBT, SFP, RENAPE, SNFCP, AFEF, SFR, and GRECCAR). Dig Liver Dis 2025; 57:669-679. [PMID: 39694751 DOI: 10.1016/j.dld.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/23/2024] [Accepted: 12/01/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND This article summarizes the French intergroup guidelines regarding rectal adenocarcinoma (RA) management published in September 2023, available on the French Society of Gastroenterology website. METHODS This work was supervised by French medical and surgical societies involved in RA management. Recommendations were rated from A to C according to the literature until September 2023. RESULTS Based on the pretreatment work-up, RA treatment was divided into four groups. T1N0 can be treated by endoscopic or surgical excision alone if there is no risk factor for lymph node involvement. For T2N0, radical surgery with total mesorectal excision is recommended, but rectal conservation is possible for small tumors (<4cm) after complete/subcomplete response following chemoradiotherapy. For T12N+ or T3+any N, total neoadjuvant treatment (TNT) followed by radical surgery is the gold standard, but rectal conservation is possible for small tumors after complete/subcomplete response following TNT. T3N2 or T+any N are an indication for TNT followed by radical surgery. Immunotherapy shows promise for dMMR/MSI RA. For metastatic tumors, recommendations are based on less robust evidence and chemotherapy plays a major role. CONCLUSION These guidelines aim at providing a personalized therapeutic strategy and are constantly being optimized. Each case should be discussed by a multidisciplinary team.
Collapse
Affiliation(s)
- Eddy Cotte
- Department of Digestive and Oncological Surgery, Lyon-Sud University Hospital, Pierre-Bénite, France.
| | - Justine Arquilliere
- Department of Digestive and Oncological Surgery, Lyon-Sud University Hospital, Pierre-Bénite, France
| | - Pascal Artru
- Department of Digestive Oncology, Jean Mermoz Private Hospital, Lyon, France
| | - Jean Baptiste Bachet
- Department of Hepato-Gastro-Enterology, Pitié-Salpêtrière Hospital Group, Assistance Publique-Hôpitaux de Paris, Pierre & Marie Curie University, Paris, France
| | - Leonor Benhaim
- Department of Visceral and Surgical Oncology, Gustave Roussy Hospital, Cancer Campus, 114 rue Edouard Vaillant, 94805 Villejuif, France
| | - Frederic Bibeau
- Department of Pathology, Besançon University Hospital, Besançon, France
| | - Niki Christou
- Department of Digestive Surgery, Limoges University Hospital, Limoges, France
| | - Thierry Conroy
- Department of Medical Oncology, Lorraine Cancer Institute, Vandoeuvre-lès-Nancy, France and Lorraine University, Inserm INSPIIRE, Nancy, France
| | - Jérome Doyen
- Department of Radiation Therapy, Antoine Lacassagne Cancer Center, University of Nice- Sophia, Nice, France
| | - Christine Hoeffel
- Department of Medical Imaging, Reims University Hospital, CRESTIC, URCA, Reims, France
| | - Nicolas Meillan
- Department of Radiation Oncology, Victor Dupouy Hospital, Argenteuil, France; Radiation Epidemiology Group, INSERM Unit 1018, Villejuif, F-94805, France
| | - Xavier Mirabel
- Academic Department of Radiation Oncology, Oscar Lambret Center, Lille, France
| | - Mathieu Pioche
- Endoscopy and Gastroenterology Unit, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | | | - Véronique Vendrely
- Department of Radiation Oncology, Haut-Lévêque Hospital, Bordeaux University, INSERM 1218-BRIC, France
| | - Florence Huguet
- Department of Radiation Oncology, Tenon Hospital, AP-HP, Sorbonne University, Paris, France
| | - Olivier Bouché
- Department of Gastroenterology and Digestive Oncology, Reims University Hospital, Reims, France
| |
Collapse
|
13
|
Wang F, Chen G, Zhang Z, Yuan Y, Wang Y, Gao Y, Sheng W, Wang Z, Li X, Yuan X, Cai S, Ren L, Liu Y, Xu J, Zhang Y, Liang H, Wang X, Zhou A, Ying J, Li G, Cai M, Ji G, Li T, Wang J, Hu H, Nan K, Wang L, Zhang S, Li J, Xu R. The Chinese Society of Clinical Oncology (CSCO): Clinical guidelines for the diagnosis and treatment of colorectal cancer, 2024 update. Cancer Commun (Lond) 2025; 45:332-379. [PMID: 39739441 PMCID: PMC11947620 DOI: 10.1002/cac2.12639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 01/02/2025] Open
Abstract
The 2024 updates of the Chinese Society of Clinical Oncology (CSCO) Clinical Guidelines for the diagnosis and treatment of colorectal cancer emphasize standardizing cancer treatment in China, highlighting the latest advancements in evidence-based medicine, healthcare resource access, and precision medicine in oncology. These updates address disparities in epidemiological trends, clinicopathological characteristics, tumor biology, treatment approaches, and drug selection for colorectal cancer patients across diverse regions and backgrounds. Key revisions include adjustments to evidence levels for intensive treatment strategies, updates to regimens for deficient mismatch repair (dMMR)/ microsatellite instability-high (MSI-H) patients, proficient mismatch repair (pMMR)/ microsatellite stability (MSS) patients who have failed standard therapies, and rectal cancer patients with low recurrence risk. Additionally, recommendations for digital rectal examination and DNA polymerase epsilon (POLE)/ DNA polymerase delta 1 (POLD1) gene mutation testing have been strengthened. The 2024 CSCO Guidelines are based on both Chinese and international clinical research, as well as expert consensus, ensuring their relevance and applicability in clinical practice, while maintaining a commitment to scientific rigor, impartiality, and timely updates.
Collapse
Affiliation(s)
- Feng Wang
- Department of Medical OncologySun Yat‐sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical SciencesGuangzhouGuangdongP. R. China
| | - Gong Chen
- Department of Colorectal SurgerySun Yat‐sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerGuangzhouGuangdongP. R. China
| | - Zhen Zhang
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiP. R. China
| | - Ying Yuan
- Department of Medical OncologyThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Yi Wang
- Department of RadiologyPeking University People's HospitalBeijingP. R. China
| | - Yuan‐Hong Gao
- Department of Radiation OncologySun Yat‐sen University Cancer Centre, The State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Weiqi Sheng
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiP. R. China
| | - Zixian Wang
- Department of Medical OncologySun Yat‐sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical SciencesGuangzhouGuangdongP. R. China
| | - Xinxiang Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiP. R. China
| | - Xianglin Yuan
- Department of OncologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Sanjun Cai
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiP. R. China
| | - Li Ren
- Department of General SurgeryZhongshan HospitalFudan UniversityShanghaiP. R. China
| | - Yunpeng Liu
- Department of Medical OncologyThe First Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Jianmin Xu
- Department of General SurgeryZhongshan HospitalFudan UniversityShanghaiP. R. China
| | - Yanqiao Zhang
- Department of OncologyHarbin Medical University Cancer HospitalHarbinHeilongjiangP. R. China
| | - Houjie Liang
- Department of OncologySouthwest HospitalThird Military Medical University (Army Medical University)ChongqingP. R. China
| | - Xicheng Wang
- Department of Gastrointestinal OncologyCancer Medical Center, Peking Union Medical College HospitalChinese Academy of Medical SciencesBeijingChina
| | - Aiping Zhou
- Department of Medical OncologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Jianming Ying
- Department of PathologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Guichao Li
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiP. R. China
| | - Muyan Cai
- Department of PathologySun Yat‐sen University Cancer Center, The State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Gang Ji
- Department of Gastrointestinal SurgeryXijing HospitalAir Force Military Medical UniversityXi'anShaanxiP. R. China
| | - Taiyuan Li
- Department of General SurgeryThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Jingyu Wang
- Department of RadiologyThe First Hospital of Jilin UniversityChangchunJilinP. R. China
| | - Hanguang Hu
- Department of Medical OncologyThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Kejun Nan
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiP. R. China
| | - Liuhong Wang
- Department of RadiologySecond Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Suzhan Zhang
- Department of Colorectal SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Jin Li
- Department of Medical OncologyShanghai GoBroad Cancer HospitalChina Pharmaceutical UniversityShanghaiP. R. China
| | - Rui‐Hua Xu
- Department of Medical OncologySun Yat‐sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat‐sen University, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical SciencesGuangzhouGuangdongP. R. China
| |
Collapse
|
14
|
Li Y, Liang F, Li Z, Zhang X, Wu A. Neoadjuvant Immunotherapy for Patients With Microsatellite Instability-High or POLE-Mutated Locally Advanced Colorectal Cancer With Bulky Tumors: New Optimization Strategy. Clin Colorectal Cancer 2025; 24:18-31.e2. [PMID: 39095269 DOI: 10.1016/j.clcc.2024.07.001] [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: 04/28/2024] [Revised: 07/01/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE To evaluate the efficacy and safety of neoadjuvant immunotherapy for patients with microsatellite instability-high (MSI-H) or DNA polymerase ε (POLE)-mutated locally advanced colorectal cancer (LACRC) with bulky tumors. PATIENTS: We retrospectively reviewed 22 consecutive patients with MSI-H or POLE-mutated LACRC with bulky tumors (>8 cm in diameter) who received preoperative programmed death-1 blockade, with or without CapOx chemotherapy. MAIN OUTCOME MEASURES: Pathological complete response (pCR), clinical complete response (cCR), toxicity, R0 resection rate, and complications were evaluated. Survival outcomes were analyzed using the Kaplan-Meier method. Multiplex immunofluorescence analysis were performed before and after treatment. RESULTS: The incidence of immune-related adverse events (irAEs) was 36.4% (8/22). Five of 22 patients presented with surgical emergencies, most commonly perforation or obstruction. The 22 patients underwent a median 4 (1-8) cycles. Two patients were evaluated as cCR and underwent a watch and wait strategy. The R0 resection rate was 100.0% (20/20) and pCR rate was 70.0% (14/20). Twelve of 14 cT4b patients (85.7%) avoided multivisceral resection, and 10 of them achieved pCR or cCR. In the two patients with POLE mutations, one each achieved pCR and cCR. No Grade III/IV postoperative complications occurred. The median follow-up was 16.0 months. Two-year event-free and overall survival for the whole cohort was both 100%. CONCLUSIONS: Preoperative immunotherapy is the optimal option for MSI-H or POLE-mutated LACRC with bulky tumors, especially cT4b. Preoperative immunotherapy in patients with T4b CRC can reduce multivisceral resection and achieve high CR rate.
Collapse
Affiliation(s)
- Yingjie Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Fei Liang
- Department of Biostatistics, Clinical Research Unit, Institute of Clinical Science Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhongwu Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaoyan Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Radiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Aiwen Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| |
Collapse
|
15
|
Raimondi A, Lonardi S, Murgioni S, Cardellino GG, Tamberi S, Strippoli A, Palermo F, De Manzoni G, Bencivenga M, Bittoni A, Chiodoni C, Lorenzini D, Todoerti K, Manca P, Sangaletti S, Prisciandaro M, Randon G, Nichetti F, Bergamo F, Brich S, Belfiore A, Bertolotti A, Stetco D, Guidi A, Torelli T, Vingiani A, Joshi RP, Khoshdeli M, Beaubier N, Stumpe MC, Nappo F, Leone AG, Pircher CC, Leoncini G, Sabella G, Airo' Farulla L, Alessi A, Morano F, Martinetti A, Niger M, Fassan M, Di Maio M, Kaneva K, Milione M, Nimeiri H, Sposito C, Agnelli L, Mazzaferro V, Di Bartolomeo M, Pietrantonio F. Tremelimumab and durvalumab as neoadjuvant or non-operative management strategy of patients with microsatellite instability-high resectable gastric or gastroesophageal junction adenocarcinoma: the INFINITY study by GONO. Ann Oncol 2025; 36:285-296. [PMID: 39637944 DOI: 10.1016/j.annonc.2024.11.016] [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: 10/15/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND In resectable gastric/gastroesophageal junction adenocarcinoma, microsatellite instability-high (MSI-H) confers improved survival, but limited benefit from chemotherapy. Immunotherapy may eliminate the need for chemotherapy or surgery. PATIENTS AND METHODS INFINITY is a multicenter, multicohort phase II trial (NCT04817826) investigating in cohort 1 the activity and safety of tremelimumab + durvalumab (T300/D) as neoadjuvant treatment of mismatch repair deficient/MSI-H, resectable gastric/gastroesophageal junction adenocarcinoma. Primary endpoint was pathologic complete response (pCR) rate; Secondary endpoints: progression-free survival (PFS), overall survival (OS), quality of life, and translational analyses. In cohort 2, the activity and safety of T300/D was explored as definitive treatment in patients achieving clinical complete response (cCR). Primary endpoint was 2-year cCR rate, and secondary endpoints were PFS, OS, quality of life, gastrectomy-free survival and translational analyses. RESULTS In cohort 1, 18 patients were recruited and 15 evaluable. pCR and major pathologic response-pCR were 60% and 80%, respectively. Since pCR rate in T4 tumors was 17%, this subgroup of patients was excluded from enrollment in cohort 2. At 28.1 months median follow-up, 24-month gastric cancer-specific PFS and OS rates were 85% and 92%, respectively. In cohort 2, 18 patients were enrolled and 17 assessable, and 13 had cCR and started non-operative management. At 11.5 months median follow-up, one patient had local regrowth and underwent salvage surgery; 12-month gastrectomy-free survival was 64.2%. CONCLUSIONS The INFINITY study provided promising activity results of a chemo-free T300/D combination regimen as preoperative treatment in mismatch repair deficient/MSI gastric/gastroesophageal junction adenocarcinoma and the first available feasibility results of a non-operative management strategy in this disease setting, worthy of further validation in larger cohorts.
Collapse
Affiliation(s)
- A Raimondi
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - S Lonardi
- Medical Oncology 3, Veneto Institute of Oncology IOV - IRCCS, Padua, USA
| | - S Murgioni
- Medical Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, USA
| | - G G Cardellino
- Department of Oncology, Presidio Ospedaliero "Santa Maria della Misericordia"-ASUFC, Udine, USA
| | - S Tamberi
- Oncology Unit, Ravenna Hospital, AUSL Romagna, Ravenna, USA
| | - A Strippoli
- Medical Oncology Unit, Policlinico Universitario A. Gemelli, Rome, USA
| | - F Palermo
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - G De Manzoni
- Department of Surgery, Azienda Ospedaliero Universitaria Integrata di Verona-Borgo Trento, Verona, USA
| | - M Bencivenga
- Department of Surgery, Azienda Ospedaliero Universitaria Integrata di Verona-Borgo Trento, Verona, USA
| | - A Bittoni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, USA
| | - C Chiodoni
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, USA
| | - D Lorenzini
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - K Todoerti
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - P Manca
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - S Sangaletti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, USA
| | - M Prisciandaro
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - G Randon
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - F Nichetti
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - F Bergamo
- Medical Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, USA
| | - S Brich
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - A Belfiore
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - A Bertolotti
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - D Stetco
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - A Guidi
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - T Torelli
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - A Vingiani
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | | | | | | | | | - F Nappo
- Medical Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, USA
| | - A G Leone
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - C C Pircher
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - G Leoncini
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - G Sabella
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - L Airo' Farulla
- Division of Nuclear Medicine, Istituto Nazionale Tumori IRCCS Milan, Milan, Italy
| | - A Alessi
- Division of Nuclear Medicine, Istituto Nazionale Tumori IRCCS Milan, Milan, Italy
| | - F Morano
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - A Martinetti
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - M Niger
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - M Fassan
- School of Medicine and Surgery, University of Padua, Padua, Italy
| | - M Di Maio
- Department of Oncology, University of Turin, at Le Molinette Hospital, Turin, Italy
| | | | - M Milione
- Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | | | - C Sposito
- Department of Oncology, University of Milan and G.I. Surgery, Istituto Nazionale Tumori IRCCS Milan, Milan, Italy
| | - L Agnelli
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA; Department of Diagnostic Innovation, Istituto Nazionale Tumori IRCCS, Milan, USA
| | - V Mazzaferro
- Department of Oncology, University of Milan and G.I. Surgery, Istituto Nazionale Tumori IRCCS Milan, Milan, Italy
| | - M Di Bartolomeo
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA
| | - F Pietrantonio
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Milan, Milan, USA.
| |
Collapse
|
16
|
Mai DVC, Drami I, Pring ET, Gould LE, Rai J, Wallace A, Hodges N, Burns EM, Jenkins JT, on behalf of the BiCyCLE Research Group. A Scoping Review of the Implications and Applications of Body Composition Assessment in Locally Advanced and Locally Recurrent Rectal Cancer. Cancers (Basel) 2025; 17:846. [PMID: 40075693 PMCID: PMC11899338 DOI: 10.3390/cancers17050846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Background: A strong body of evidence exists demonstrating deleterious relationships between abnormal body composition (BC) and outcomes in non-complex colorectal cancer. Complex rectal cancer (RC) includes locally advanced and locally recurrent tumours. This scoping review aims to summarise the current evidence examining BC in complex RC. Methods: A literature search was performed on Ovid MEDLINE, EMBASE, and Cochrane databases. Original studies examining BC in adult patients with complex RC were included. Two authors undertook screening and full-text reviews. Results: Thirty-five studies were included. Muscle quantity was the most commonly studied BC metric, with sarcopenia appearing to predict mortality, recurrence, neoadjuvant therapy outcomes, and postoperative complications. In particular, 10 studies examined relationships between BC and neoadjuvant therapy response, with six showing a significant association with sarcopenia. Only one study examined interventions for improving BC in patients with complex RC, and only one study specifically examined patients undergoing pelvic exenteration. Marked variation was also observed in terms of how BC was quantified, both in terms of anatomical location and how cut-off values were defined. Conclusions: Sarcopenia appears to predict mortality and recurrence in complex RC. An opportunity exists for a meta-analysis examining poorer BC and neoadjuvant therapy outcomes. There is a paucity of studies examining interventions for poor BC. Further research examining BC specifically in patients undergoing pelvic exenteration surgery is also lacking. Pitfalls identified include variances in how BC is measured on computed tomography and whether external cut-off values for muscle and adipose tissue are appropriate for a particular study population.
Collapse
Affiliation(s)
- Dinh Van Chi Mai
- St Mark’s Hospital and Academic Institute, St Mark’s The National Bowel Hospital, London HA1 3UJ, UK
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| | - Ioanna Drami
- St Mark’s Hospital and Academic Institute, St Mark’s The National Bowel Hospital, London HA1 3UJ, UK
- Department of Digestion, and Reproduction, Imperial College London, London W12 0NN, UK
| | - Edward T. Pring
- St Mark’s Hospital and Academic Institute, St Mark’s The National Bowel Hospital, London HA1 3UJ, UK
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| | - Laura E. Gould
- St Mark’s Hospital and Academic Institute, St Mark’s The National Bowel Hospital, London HA1 3UJ, UK
- School of Cancer Sciences, College of Veterinary & Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Jason Rai
- St Mark’s Hospital and Academic Institute, St Mark’s The National Bowel Hospital, London HA1 3UJ, UK
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| | - Alison Wallace
- St Mark’s Hospital and Academic Institute, St Mark’s The National Bowel Hospital, London HA1 3UJ, UK
- School of Cancer Sciences, College of Veterinary & Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Nicola Hodges
- St Mark’s Hospital and Academic Institute, St Mark’s The National Bowel Hospital, London HA1 3UJ, UK
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| | - Elaine M. Burns
- St Mark’s Hospital and Academic Institute, St Mark’s The National Bowel Hospital, London HA1 3UJ, UK
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| | - John T. Jenkins
- St Mark’s Hospital and Academic Institute, St Mark’s The National Bowel Hospital, London HA1 3UJ, UK
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| | | |
Collapse
|
17
|
Zhou J, Zhang X, Liu Q, Li Y, Wu G, Fu W, Yao H, Wang Z, Xue H, Xu T, Chen W, Lu J, Zhang G, Wu B, An Y, Qiu X, Xiao Y, Lin G. Rationale and design of a multicentre randomised controlled trial on circulating tumour DNA-guided neoadjuvant treatment strategy for locally advanced rectal cancer (CINTS-R). BMJ Open 2025; 15:e090765. [PMID: 39894522 PMCID: PMC11792281 DOI: 10.1136/bmjopen-2024-090765] [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: 07/03/2024] [Accepted: 01/10/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND The traditional neoadjuvant chemoradiotherapy (nCRT) combined with total mesorectal excision has been widely accepted as the standard treatment for patients with locally advanced rectal cancer (LARC). New strategies such as total neoadjuvant therapy (TNT) and neoadjuvant immunotherapy have shown great promise in certain patient populations. Currently, there is an urgent need to stratify patients before treatment to adopt the appropriate neoadjuvant strategies. Our previous study has shown that circulating tumour DNA (ctDNA) effectively reflects tumour burden and genetic characteristics and has significant predictive value for tumour recurrence, demonstrating great potential in guiding the choice of neoadjuvant strategies. METHODS AND ANALYSIS The CINTS-R trial is a multicentre, open-label, randomised controlled trial designed to evaluate the efficacy and safety of a ctDNA-guided neoadjuvant treatment strategy compared with conventional neoadjuvant therapy regime in patients with LARC. The trial will enrol 470 patients diagnosed with LARC (staged cT3-4N0 or cTanyN1-2) with tumours located ≤12 cm from the anal verge across seven centres in China. Patients will be randomly assigned in a 2:1 ratio to the experimental group or the control group. Patients in the experimental group will receive different intensities of neoadjuvant chemoradiotherapy (TNT or modified nCRT) or neoadjuvant immunotherapy based on the molecular features of the tumour, baseline ctDNA concentration and changes in ctDNA status early in treatment. Patients in the control group will receive modified nCRT. The primary endpoint is the 2-year disease-related treatment failure rate. The secondary endpoints include time to recurrence, 2-year overall survival, 2-year disease-free survival, clinical complete response (cCR) rate, near cCR rate and pathologically complete response rate, pathological tumour regression grade and quality of life. ETHICS AND DISSEMINATION This protocol has been approved by the ethics committee of Peking Union Medical College Hospital, with approval number I-23PJ157, and by the institutional review boards of all the participating centres. All data will be collected and stored in a specially designed database. The results of our trial will be disseminated through peer-reviewed publications and presented at national and international academic conferences. TRIAL REGISTRATION NUMBER This trial is registered on ClinicalTrials.gov and the registration ID is NCT05601505.
Collapse
Affiliation(s)
- Jiaolin Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Xiao Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Qian Liu
- Department of Colorectal Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Yongheng Li
- Department of Radiation Oncology, Peking University Cancer Hospital, Beijing, China
| | - Guoju Wu
- Department of General Surgery, Beijing Hospital, Beijing, China
| | - Wei Fu
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Hongwei Yao
- Department of General Surgery, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Zhenjun Wang
- Department of General Surgery, Beijing Chao-Yang Hospital, Beijing, China
| | - Huadan Xue
- Department of Radiology, Peking Union Medical College Hospital, Beijing, China
| | - Tao Xu
- Department of Epidemiology and Biostatistics Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College School of Basic Medicine, Beijing, China
| | - Weijie Chen
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Junyang Lu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Guannan Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Bin Wu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Yang An
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Xiaoyuan Qiu
- Peking Union Medical College Hospital, Beijing, China
| | - Yi Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Guole Lin
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
18
|
Ke H, Li P, Li Z, Zeng X, Zhang C, Luo S, Chen X, Zhou X, Dong S, Chen S, Huang J, Yuan M, Yu R, Ye S, Hu T, Tang Z, Liu D, Wu K, Wu X, Lan P. Immune profiling of the macroenvironment in colorectal cancer unveils systemic dysfunction and plasticity of immune cells. Clin Transl Med 2025; 15:e70175. [PMID: 39934971 PMCID: PMC11813809 DOI: 10.1002/ctm2.70175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/01/2024] [Accepted: 12/29/2024] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Tumour immune macroenvironment is comprised of tumour and surrounding organs responding to tumourigenesis and immunotherapy. The lack of comprehensive analytical methods hinders its application for prediction of survival and treatment response in colorectal cancer (CRC) patients. METHODS Cytometry by time-of-flight (CyTOF) and RNA-seq was applied to characterise immune cell heterogeneity in a discovery cohort including tumour, blood and intestinal architecture comprising epithelium, lamina propria, submucosa, muscularis propria of normal bowel and tumour-adjacent bowel tissues. Immunoprofiling was also validated by a validation cohort using single-cell RNA sequencing, spatial transcription, CyTOF and multiplex immunofluorescent staining. RESULTS Based on cell phenotype and transcription, we identify distinct immunotypes in the CRC macroenvironment including blood, tumour and different intestinal architecture, showing disturbed immune cell compositions, increasing expression of immunosuppressive markers and cell-cell interactions contributing to immunosuppressive regulation. Furthermore, we evaluate immune macroenvironment influencing factors including tertiary lymphoid structures (TLSs), consensus molecular subtypes (CMSs) and immune checkpoint inhibitors (ICIs). TLS presence fuels anti-tumour immunity by promoting CD8+ T cell infiltration and altering activation or suppression of T cell systematically. TLS presence correlates with patient survival, intrinsic CMS and therapeutic efficacy of ICI. PD-1 and CD69 expressed in effector memory CD8+ T cells from blood can predict TLS presence in the CRC macroenvironment, serving as potential biomarkers for stratifying CRC patients into immunotherapy. CONCLUSIONS Our findings provide insights into the CRC immune macroenvironment, highlighting immune cell suppression and activation in tumourigenesis. Our study illustrates the potential utility of blood for predicting immunotherapy response. KEY POINTS Distinct immunotypes are identified in the CRC macroenvironment. TLS and immunotherapy exert influence on the immune macroenvironment. TLS presence correlates with patient survival, CMS and therapeutic efficacy of ICI. PD-1 and CD69 expressed in CD8+ Tem from blood can predict TLS presence in the CRC macroenvironment.
Collapse
Affiliation(s)
- Haoxian Ke
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Gastrointestinal Surgery, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Peisi Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Zhihao Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Colorectal Surgery, Department of General SurgeryThe Sixth Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Xian Zeng
- Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Chi Zhang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Colorectal Surgery, Department of General SurgeryThe Sixth Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Shuzhen Luo
- HIM‐BGI Omics Center, Hangzhou Institute of Medicine (HIM)Chinese Academy of Sciences, BGI ResearchHangzhouChina
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of GenomicsBGI ResearchShenzhenChina
- BGI GenomicsHarbinChina
| | - Xiaofang Chen
- HIM‐BGI Omics Center, Hangzhou Institute of Medicine (HIM)Chinese Academy of Sciences, BGI ResearchHangzhouChina
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of GenomicsBGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Xinlan Zhou
- HIM‐BGI Omics Center, Hangzhou Institute of Medicine (HIM)Chinese Academy of Sciences, BGI ResearchHangzhouChina
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of GenomicsBGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Shichen Dong
- BGI ResearchShenzhenChina
- Shenzhen Key Laboratory of Single‐Cell OmicsBGI ResearchShenzhenChina
| | - Shaopeng Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Colorectal Surgery, Department of General SurgeryThe Sixth Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Junfeng Huang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Colorectal Surgery, Department of General SurgeryThe Sixth Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Ming Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Colorectal Surgery, Department of General SurgeryThe Sixth Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Runfeng Yu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Colorectal Surgery, Department of General SurgeryThe Sixth Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Shubiao Ye
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Tuo Hu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Colorectal Surgery, Department of General SurgeryThe Sixth Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Zhonghui Tang
- Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Dongbin Liu
- HIM‐BGI Omics Center, Hangzhou Institute of Medicine (HIM)Chinese Academy of Sciences, BGI ResearchHangzhouChina
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of GenomicsBGI ResearchShenzhenChina
- BGI GenomicsHarbinChina
| | - Kui Wu
- HIM‐BGI Omics Center, Hangzhou Institute of Medicine (HIM)Chinese Academy of Sciences, BGI ResearchHangzhouChina
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of GenomicsBGI ResearchShenzhenChina
- BGI GenomicsHarbinChina
| | - Xianrui Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Gastrointestinal Surgery, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Ping Lan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Gastrointestinal Surgery, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Colorectal Surgery, Department of General SurgeryThe Sixth Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
19
|
Ji P, Chen T, Li C, Zhang J, Li X, Zhu H. Comprehensive review of signaling pathways and therapeutic targets in gastrointestinal cancers. Crit Rev Oncol Hematol 2025; 206:104586. [PMID: 39653094 DOI: 10.1016/j.critrevonc.2024.104586] [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: 05/31/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
Targeted therapy, the milestone in the development of human medicine, originated in 2004 when the FDA approved the first targeted agent bevacizumab for colorectal cancer treatment. This new development has resulted from drug developers moving beyond traditional chemotherapy, and several trials have popped up in the last two decades with an unprecedented speed. Specifically, EGF/EGFR, VEGF/VEGFR, HGF/c-MET, and Claudin 18.2 therapeutic targets have been developed in recent years. Some targets previously thought to be undruggable are now being newly explored, such as the RAS site. However, the efficacy of targeted therapy is extremely variable, especially with the emergence of new drugs and the innovative use of traditional targets for other tumors in recent years. Accordingly, this review provides an overview of the major signaling pathway mechanisms and recent advances in targeted therapy for gastrointestinal cancers, as well as future perspectives.
Collapse
Affiliation(s)
- Pengfei Ji
- Department of Thoracic Surgery, West China Hospital, Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China
| | - Tingting Chen
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Chao Li
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Jinyuan Zhang
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Xiao Li
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
20
|
Liu B, Xie Y, Zhang Y, Tang G, Lin J, Yuan Z, Liu X, Wang X, Huang M, Luo Y, Yu H. Spatial deconvolution from bulk DNA methylation profiles determines intratumoral epigenetic heterogeneity. Cell Biosci 2025; 15:7. [PMID: 39844296 PMCID: PMC11756021 DOI: 10.1186/s13578-024-01337-y] [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: 09/30/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Intratumoral heterogeneity emerges from accumulating genetic and epigenetic changes during tumorigenesis, which may contribute to therapeutic failure and drug resistance. However, the lack of a quick and convenient approach to determine the intratumoral epigenetic heterogeneity (eITH) limit the application of eITH in clinical settings. Here, we aimed to develop a tool that can evaluate the eITH using the DNA methylation profiles from bulk tumors. METHODS Genomic DNA of three laser micro-dissected tumor regions, including digestive tract surface, central bulk, and invasive front, was extracted from formalin-fixed paraffin-embedded sections of colorectal cancer patients. The genome-wide methylation profiles were generated with methylation array. The most variable methylated probes were selected to construct a DNA methylation-based heterogeneity (MeHEG) estimation tool that can deconvolve the proportion of each reference tumor region with the support vector machine model-based method. A PCR-based assay for quantitative analysis of DNA methylation (QASM) was developed to specifically determine the methylation status of each CpG in MeHEG assay at single-base resolution to realize fast evaluation of epigenetic heterogeneity. RESULTS In the discovery set with 79 patients, the differentially methylated CpGs among the three tumor regions were found. The 7 most representative CpGs were identified and subsequently selected to develop the MeHEG algorithm. We validated its performance of deconvolution of tumor regions in an independent cohort. In addition, we showed the significant association of MeHEG-based epigenetic heterogeneity with the genomic heterogeneity in mutation and copy number variation in our in-house and TCGA cohorts. Besides, we found that the patients with higher MeHEG score had worse disease-free and overall survival outcomes. Finally, we found dynamic change of epigenetic heterogeneity based on MeHEG score in cancer cells under the treatment of therapeutic drugs. CONCLUSION By developing a 7-loci panel using a machine learning approach combined with the QASM assay for PCR-based application, we present a valuable method for evaluating intratumoral heterogeneity. The MeHEG algorithm offers novel insights into tumor heterogeneity from an epigenetic perspective, potentially enriching current knowledge of tumor complexity and providing a new tool for clinical and research applications in cancer biology.
Collapse
Affiliation(s)
- Binbin Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
| | - Yumo Xie
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
| | - Yu Zhang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
| | - Guannan Tang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
| | - Jinxin Lin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
| | - Ze Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
| | - Xiaoxia Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Guangzhou, Guangdong, China
- Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaolin Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Guangzhou, Guangdong, China
- Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Meijin Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Guangzhou, Guangdong, China
- Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanxin Luo
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Guangzhou, Guangdong, China
- Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huichuan Yu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China.
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Guangzhou, Guangdong, China.
- Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
21
|
Chen X, Chen J, Xu L, Lin D, Hong X, Peng J, He X, Hu J. DMMR status and synchronous lesions predicts metachronous lesions after curative resection for rectal cancer. Front Surg 2025; 12:1510400. [PMID: 39906700 PMCID: PMC11790672 DOI: 10.3389/fsurg.2025.1510400] [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: 10/12/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025] Open
Abstract
Background There are no established standard colonoscopy surveillance protocols for patients after curative rectal cancer resection. We investigated the predictive factors for colorectal neoplasms during surveillance colonoscopies to identify patients who are at risk of developing metachronous neoplasms in the residual colorectum. Methods This was a single-center, retrospective study that included patients with diagnosis of rectal carcinoma who had undergone curative resection from October 2012 to June 2018. Clinicopathological variables were analyzed by logistic regression analysis to identify risk factors independently associated with metachronous neoplasms in patients that underwent curative rectal cancer surgery. Results In all, 554 patients were included in the analysis. Deficient mismatch repair (dMMR) status was recorded in 20 (3.6%) patients. At the surveillance colonoscopies, 118 patients (21.3%) had metachronous neoplasms while 169 patients (30.5%) had metachronous polyps. The median time interval between index colonoscopy and the last surveillance colonoscopy was 736.5 (476.75-1,082.25) days. Univariable and multivariable analysis showed dMMR status, synchronous adenomas/polyps, surveillance time > 3, and longer surveillance period patients were significant risk factors for development of metachronous lesions; in subgroup analysis, we also found that among rectal cancer patients with synchronous adenomas, adenomas located in the left colon and rectum, and longer surveillance period were independent risk factors for detecting metachronous adenomas. Conclusions This study underscored the importance of extended follow-up protocols and targeted surveillance for identifying and managing metachronous lesions in dMMR rectal cancer patients, especially with synchronous adenomas. Further prospective, multicenter studies are needed to validate these results.
Collapse
Affiliation(s)
- Xijie Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Gastric Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Junguo Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liang Xu
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dezheng Lin
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Endoscopic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoling Hong
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Endoscopic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Junsheng Peng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Gastric Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaowen He
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiancong Hu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Endoscopic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
22
|
Wang H, Zhai M, Li M, Han C, Liu L, Huang C, Zhao L, Yu D, Tao K, Ren J, Lin Z, Zhang T. Phenotypic plasticity and increased infiltration of peripheral blood-derived TREM1 + mono-macrophages following radiotherapy in rectal cancer. Cell Rep Med 2025; 6:101887. [PMID: 39793571 PMCID: PMC11866444 DOI: 10.1016/j.xcrm.2024.101887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/29/2024] [Accepted: 12/05/2024] [Indexed: 01/13/2025]
Abstract
In our previously reported phase 2 and phase 3 studies, the combination of short-course radiotherapy and neoadjuvant immunochemotherapy (SIC) is established as effective cancer therapies for locally advanced rectal cancer (LARC). Here, we apply multi-omic analyses to paired pre- and post-treatment LARC specimens undergoing SIC. The peripheral blood-derived TREM1+ mono-macrophage subsets that display a pro-inflammatory phenotype are identified and correlate with complete response to SIC. Mechanically, ionizing radiation (IR) induces peripheral TREM1+ mono-macrophage expansion in tumors. Following IR, the loss of TREM1 in mono-macrophages undermines antitumor immunity by altering mono-macrophage differentiation and inhibiting CD8+ T cell infiltration and activation. The TREM1+ mono-macrophage response may rely on activation of key inflammatory pathways, including nuclear factor κB (NF-κB) signaling and Toll-like receptor pathway. Pharmacological inhibition of TREM1 signaling abolishes IR-induced immunoactivation and reduces combined IR and/or anti-PD-1 treatment. Thus, we establish a crucial role of a mono-macrophage state in mediating effective cancer therapy.
Collapse
Affiliation(s)
- Haihong Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Menglan Zhai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Mingjie Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Chaoqun Han
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lichao Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Chuying Huang
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, China
| | - Lei Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Dandan Yu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Zhenyu Lin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China.
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China.
| |
Collapse
|
23
|
Ai L, Li Q, Zhang S, Dong Y, Yang M, Li J, Pan Y, Yuan Y, Yi S, Wang J, Cheng Y, Feng J, Gao S, Wang X, Qu S, Zhang X, Lu J, Xiu P, Wang S, Yang X, Yu Y, Liu T. Famitinib plus camrelizumab in patients with advanced colorectal cancer: Data from a multicenter, basket study. Innovation (N Y) 2025; 6:100745. [PMID: 39872476 PMCID: PMC11763884 DOI: 10.1016/j.xinn.2024.100745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 11/26/2024] [Indexed: 01/30/2025] Open
Abstract
Concurrent inhibition of angiogenesis and immune checkpoints represents a potent therapeutic approach. We conducted a phase 2, multicenter, basket study to assess the efficacy and safety of combination therapy of famitinib (anti-angiogenic agent) plus camrelizumab (PD-1 antagonist) in patients with metastatic solid tumors across 11 cohorts (this study was registered at Clinicaltrials.gov [NCT04346381]). This report focuses on the cohort of patients with metastatic or advanced colorectal cancer. Eligible patients, who had previously received ≥2 lines of systemic treatments for their metastatic disease, were treated with famitinib (20 mg once daily) in combination with camrelizumab (200 mg intravenously every 3 weeks). The primary endpoint was the objective response rate, with secondary endpoints encompassing progression-free survival, overall survival, duration of response, safety and exploratory biomarkers. A total of 44 patients were enrolled and treated. With a median follow-up time of 9.46 months (range 2.0-22.5 months), objective responses were observed in 6 patients (13.6%; 95% confidence interval [CI], 5.2%-27.4%), all of whom had rectal cancer. The median duration of response is 6.2 months (95% CI, 2.3-10.6 months). Median progression-free survival was 3.3 months (95% CI, 2.1-4.1 months), and median overall survival was 10.9 months (95% CI, 7.6-15.2 months). Among the 44 patients, 29 (65.9%) experienced grade 3 or 4 treatment-related adverse events, predominantly hypertension and proteinuria. In conclusion, the combination of famitinib and camrelizumab demonstrates promising antitumor activity with a manageable safety profile in metastatic colorectal cancer patients. Further research is warranted to confirm and extend these findings.
Collapse
Affiliation(s)
- Luoyan Ai
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Qian Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Shilong Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Yu Dong
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Mudan Yang
- The Second Ward of Gastroenterology Department, Shanxi Province Cancer Hospital, Taiyuan 030000, China
| | - Jin Li
- Department of Oncology, Shanghai East Hospital, Shanghai 200120, China
| | - Yueyin Pan
- Oncology Chemotherapy Department, the First Affiliated Hospital of USTC Hospital/Anhui Provicinal Hospital, Hefei 230001, China
| | - Ying Yuan
- Department of Medical Oncology, the Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310022, China
| | - Shanyong Yi
- Department of Medical Oncology, Zhengzhou Central Hospital-Department of Medical Oncology, Zhengzhou 450000, China
| | - Junsheng Wang
- Department of Medical Oncology, AnYang Cancer Hospital, Anyang 455000, China
| | - Ying Cheng
- Department of Medical Oncology, JiLin Cancer Hospital, Changchun 130000, China
| | - Jifeng Feng
- Department of Medical Oncology, Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Shegan Gao
- Department of Medical Oncology, The First Affiliated Hospital of Henan University of Science & Technology-Cancer Hospital, Luoyang 471003, China
| | - Xicheng Wang
- Department of Oncology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Song Qu
- Department of Radiotherapy, Guangxi Medical University Affiliated Tumor Hospital, Nanning 530021, China
| | - Xizhi Zhang
- Oncology Department, Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Jin Lu
- Department of Abdominal Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to School of Medicine, UESTC, Chengdu 610000, China
| | - Peng Xiu
- Clinical Research & Development, Jiangsu Hengrui Medicine Co., Ltd, Shanghai 201203, China
| | - Shuni Wang
- Clinical Research & Development, Jiangsu Hengrui Medicine Co., Ltd, Shanghai 201203, China
| | - Xinfeng Yang
- Clinical Research & Development, Jiangsu Hengrui Medicine Co., Ltd, Shanghai 201203, China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| |
Collapse
|
24
|
Wang W, Mei Z, Chen Y, Jiang J, Qu Y, Saifuding K, Zhou N, Bulibu G, Tang Y, Zhai X, Jiang Z. Immune checkpoint inhibitors for patients with mismatch repair deficient or microsatellite instability-high advanced cancers: a meta-analysis of phase I-III clinical trials. Int J Surg 2025; 111:1357-1372. [PMID: 39166943 PMCID: PMC11745646 DOI: 10.1097/js9.0000000000002007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Mismatch repair deficient (dMMR) and microsatellite instability-high (MSI-H) cancers are associated with an increased number of somatic mutations, which can render tumors more susceptible to immune checkpoint blockade. However, a comprehensive evaluation of the efficacy profile of immune checkpoint inhibitors in this patient population across multiple cancer types is lacking. This study aims to address this knowledge gap by synthesizing data from phase I-III clinical trials. METHODS A systematic search was conducted in PubMed, Embase, the Cochrane Central Register of Controlled Trials, and Google Scholar from inception until June 2024. Eligible studies included randomized controlled trials (RCTs), nonrandomized comparative studies, and single-arm trials investigating immune checkpoint inhibitors in patients with dMMR/MSI-H advanced cancers. The primary outcome was objective response rate (ORR), and the secondary outcomes included disease control rate (DCR), 1-year, 2-year, and 3-year overall survival (OS) and progression-free survival (PFS) rates. Subgroup analyses were conducted for the primary outcome stratified by major study characteristics. RESULTS Of the 10 802 identified studies, 19 trials in 25 studies totaling 2052 participants met the inclusion criteria and were included in the meta-analysis. The pooled ORR was 41.7% (95% CI, 35.7-47.7%). The pooled DCR was 68.9% (95% CI, 62.2-75.7%). The pooled 12-month, 24-month, and 36-month OS rates were 29.1% (95% CI, 19.9-38.3%), 35.8% (95% CI, 23.6-48.0%), and 35.8% (95% CI, 23.6-48.0%), respectively. The pooled 12-month, 24-month, and 36-month PFS rates were 46.4% (95% CI, 39.1-53.8%), 67.0% (95% CI, 55.2-78.8%), and 63.1% (95% CI, 37.3-88.9%), respectively. CONCLUSIONS The study establishes the therapeutic potential of immune checkpoint inhibitors in dMMR/MSI-H advanced cancers, highlighting the importance of MSI status in this context. Further, head-to-head comparisons are needed to conclusively determine MSI's predictive power relative to proficient mismatch repair/microsatellite stable (pMMR/MSS) tumors.
Collapse
Affiliation(s)
- Wei Wang
- Department of Digestive Internal Medicine, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xin Jiang Province
| | - Zubing Mei
- Department of Anorectal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine
- Anorectal Disease Institute of Shuguang Hospital
| | - Yajie Chen
- Department of Digestive Internal Medicine, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xin Jiang Province
| | - Jian Jiang
- Department of Digestive Internal Medicine, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xin Jiang Province
| | - Yanli Qu
- Department of Digestive Internal Medicine, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xin Jiang Province
| | - Keyoumu Saifuding
- Department of Digestive Internal Medicine, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xin Jiang Province
| | - Ning Zhou
- Department of Digestive Internal Medicine, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xin Jiang Province
| | - Gilisihan Bulibu
- Department of Digestive Internal Medicine, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xin Jiang Province
| | - Yong Tang
- Department of Digestive Internal Medicine, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xin Jiang Province
| | - Xinyu Zhai
- Department of Urology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine Shanghai
| | - Zhi Jiang
- Department of Perioperative Research Center of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, People’s Republic of China
| |
Collapse
|
25
|
Orhan A, Justesen TF, Raskov H, Qvortrup C, Gögenur I. Introducing Neoadjuvant Immunotherapy for Colorectal Cancer: Advancing the Frontier. Ann Surg 2025; 281:95-104. [PMID: 39005208 DOI: 10.1097/sla.0000000000006443] [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: 07/16/2024]
Abstract
OBJECTIVE To give surgeons a review of the current and future use of neoadjuvant immunotherapy in patients with localized colorectal cancer (CRC). BACKGROUND Immunotherapy has revolutionized the standard of care in oncology and improved survival outcomes in several cancers. However, the applicability of immunotherapy is still an ongoing challenge. Some cancer types are less responsive to immunotherapy, and the heterogeneity in responses within cancer types is poorly understood. Clinical characteristics of the patient, the timing of immunotherapy in relation to surgery, diversities in the immune responses, clonal heterogeneity, different features of the tumor microenvironment, and genetic alterations are some factors among many that may influence the efficacy of immunotherapy. RESULTS In this narrative review, we describe the major types of immunotherapy used to treat localized CRC. Furthermore, we discuss the prediction of response to immunotherapy in relation to biomarkers and radiologic assessment. Finally, we consider the future perspectives of clinical implications and response patterns, as well as the potential and challenges of neoadjuvant immunotherapy in localized CRC. CONCLUSIONS Establishing mismatch repair (MMR) status at the time of diagnosis is central to the potential use of neoadjuvant immunotherapy, in particular immune checkpoint inhibitors, in localized CRC. To date, efficacy is primarily seen in patients with deficient MMR status and polymerase epsilon mutations, although a small group of patients with proficient MMR does respond. In conclusion, neoadjuvant immunotherapy shows promising complete response rates, which may open a future avenue of an organ-sparing watch-and-wait approach for a group of patients.
Collapse
Affiliation(s)
- Adile Orhan
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Tobias F Justesen
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Hans Raskov
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Camilla Qvortrup
- Department of Clinical Oncology, Rigshospitalet, Copenhagen, Denmark
| | - Ismail Gögenur
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Yu Y, Huang L, Yan R, Jiang M, Li SJ, Fan WD. The efficacy and safety of neoadjuvant treatment with the PD-1 inhibitor for locally advanced colorectal cancer: a meta-analysis. Front Oncol 2024; 14:1416943. [PMID: 39687895 PMCID: PMC11646803 DOI: 10.3389/fonc.2024.1416943] [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: 04/13/2024] [Accepted: 11/05/2024] [Indexed: 12/18/2024] Open
Abstract
Objective To systematically evaluate the efficacy and safety of PD-1 inhibitors in neoadjuvant therapy for locally advanced colorectal cancer (LACRC). Method Retrieved from PubMed, Embase, and the Cochrane Library, all relevant studies about PD-1 inhibitors for neoadjuvant treatment of LACRC were collected from inception to 31 December 2023. The efficacy was assessed by the rate of pathological complete response (PCR), clinical complete response (CCR), and major pathological response (MPR), and the safety was evaluated by the incidence of all adverse effects (TRAEs). Subgroup analysis was conducted by experimental design, types of PD-1 inhibitors, and disease types. Result A total of 803 patients were included in 21 studies. The results of the meta-analysis showed that the PCR rate of PD-1 inhibitors in the treatment of LACRC was 54% (95% CI: 43%-65%, P<0.05); the CCR of anti-PD-1 was 40% (95% CI: 26%-54%, P<0.05); the MPR was 66% (95% CI: 56%-76%, P<0.05); and the irAEs was 27% (95% CI: 17%-37%, P<0.05). Subgroup analysis showed that the PCRs in prospective studies and retrospective studies were 49% (95% CI: 32%-66%, P<0.05) and 57% (95% CI: 42%-73%, P<0.05), respectively. Among the 803 patients, 619 (77%) were diagnosed with rectal cancer (RC), and the PCR and MPR were 49% and 65%, respectively; 184 (23%) were diagnosed with colorectal cancer (CRC), and the PCR and MPR were both 67%. In our meta-analysis, types of PD-1 inhibitors, including sintilimab, toripalimab, camrelizumab, avelumab, pembrolizumab, and tislelizumab, and patients who received PD-1 inhibitors alone or in combination achieved good PCR rates. Conclusion Neoadjuvant therapy combined with a PD-1 inhibitor has a favorable PCR and relatively low incidences of irAEs for patients with LACRC, suggesting that this regimen including a PD-1 inhibitor is significantly effective and sufficiently safe.
Collapse
Affiliation(s)
- Yan Yu
- West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
- West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lin Huang
- West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
- West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Yan
- West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
- West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Jiang
- West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
- West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shuang-Jiao Li
- West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
- West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wang-Dong Fan
- West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
- West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
27
|
Williams CJM, Peddle AM, Kasi PM, Seligmann JF, Roxburgh CS, Middleton GW, Tejpar S. Neoadjuvant immunotherapy for dMMR and pMMR colorectal cancers: therapeutic strategies and putative biomarkers of response. Nat Rev Clin Oncol 2024; 21:839-851. [PMID: 39317818 DOI: 10.1038/s41571-024-00943-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 09/26/2024]
Abstract
Approximately 15% of locally advanced colorectal cancers (CRC) have DNA mismatch repair deficiency (dMMR), resulting in high microsatellite instability and a high tumour mutational burden. These cancers are frequently sensitive to therapy with immune-checkpoint inhibitors (ICIs) in the metastatic setting. This sensitivity seems to be even more pronounced in locally advanced disease, and organ preservation has become a realistic aim in ongoing clinical trials involving patients with dMMR rectal cancer. By contrast, metastatic CRCs with proficient DNA mismatch repair (pMMR) are generally resistant to ICIs, although a proportion of locally advanced pMMR tumours seem to have a high degree of sensitivity to ICIs. In this Review, we describe the current and emerging clinical evidence supporting the use of neoadjuvant ICIs in patients with dMMR and pMMR CRC, and the potential advantages (based on a biological rationale) of such an approach. We discuss how neoadjuvant 'window-of-opportunity' trials are being leveraged to progress biomarker discovery and we provide an overview of potential predictive biomarkers of response to ICIs, exploring the challenges faced when evaluating such biomarkers in biopsy-derived samples. Lastly, we describe how these discoveries might be used to drive a rational approach to trialling novel immunotherapeutic strategies in patients with pMMR CRC, with the ultimate aim of disease eradication and the generation of long-term immunosurveillance.
Collapse
Affiliation(s)
| | | | - Pashtoon M Kasi
- Department of Gastrointestinal Oncology, City of Hope Orange County Lennar Foundation Cancer Center, Irvine, CA, USA
| | - Jenny F Seligmann
- Division of Oncology, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | | | - Gary W Middleton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | |
Collapse
|
28
|
Justesen TF, Orhan A, Rosen AW, Gögenur M, Gögenur I. Mismatch Repair Status and Surgical Outcomes in Localized Colorectal Cancer: A Nationwide Cohort Study. ANNALS OF SURGERY OPEN 2024; 5:e499. [PMID: 39711680 PMCID: PMC11661751 DOI: 10.1097/as9.0000000000000499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 12/24/2024] Open
Abstract
Objective This study examined the association between deficient mismatch repair (dMMR) versus proficient MMR (pMMR) status and overall survival and disease-free survival in patients with localized colorectal cancer. Background Several distinctions exist between patients with dMMR and pMMR colorectal cancer. However, the impact on prognosis is yet to be investigated in large-scale cohort studies. Methods In this cohort study, we included patients who underwent curative-intent surgery for localized colorectal cancer between 2009 and 2020. Patients were identified in the Danish Colorectal Cancer Group database and patient-level data were extracted from 6 registry databases. After inclusion, patients with dMMR status were matched 1:1 to patients with pMMR status using an estimated propensity score. Results After matching, 5994 patients were included. The patients had a median age of 74 years and a median follow-up of 4.1 years. There was no significant association between mismatch repair (MMR) status and overall survival (hazard ratio, 0.91; 95% confidence interval [CI], 0.81-1.03) or disease-free survival (hazard ratio, 0.89; 95% CI, 0.78-1.01). However, the restricted 5-year mean disease-free survival time, calculated due to violation of the proportional hazards assumption, showed a significant absolute difference of 0.13 years (95% CI, 0.03-0.23; P = 0.01) in favor of the dMMR group. Conclusions No significant association with overall survival was found according to MMR status. dMMR status was, however, found to be associated with marginally improved disease-free survival compared to pMMR status in patients with localized colorectal cancer undergoing curative-intent surgery.
Collapse
Affiliation(s)
- Tobias Freyberg Justesen
- From the Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Adile Orhan
- From the Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Andreas Weinberger Rosen
- From the Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Mikail Gögenur
- From the Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Ismail Gögenur
- From the Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
Wang C, Liang N, Qiao L, Wu Y, Zhang J, Zhang Y. Clinical features and prognosis analysis of stage III/IV patients with lung cancer after treatment with toripalimab: A real-world retrospective. J Cancer Res Ther 2024; 20:2021-2028. [PMID: 39792412 DOI: 10.4103/jcrt.jcrt_500_24] [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: 03/13/2024] [Accepted: 07/01/2024] [Indexed: 01/12/2025]
Abstract
AIM Toripalimab is the first antitumor programmed cell death protein 1 (PD-1) antibody approved in China. For better patient management, it is important to understand the real-world outcomes of toripalimab in treating patients with lung cancer in the real world outside of clinical trials to improve patient care. METHODS We retrospectively examined the clinical data of 80 patients with lung cancer who received the PD-1 inhibitor (toripalimab). The Chi-square test was performed to identify clinical factors associated with the advancement of the disease. Multivariate Cox regression analysis was used to screen prognostic variables linked to real-world progression-free survival (PFS) and overall survival (OS). OS and PFS were calculated using the Kaplan-Meier method, and the comparisons were determined using the log-rank test, and continuous and categorical variables were explained using median and percentage, respectively. RESULT The median OS of the estimated 80 patients was 15.85 months (95% confidence interval [CI]: 14.103-17.949 months), and the estimated PFS was 5.650 months (95% CI: 7.226-11.264 months). The longer OS and PFS correlate with the patient's staging and number of treatment lines. The PD-1 drug gave stage III patients a significantly longer PFS and OS compared to stage IV patients (PFS: 14.65 vs. 6.68, P = 0.004; OS: 21.1 vs. 13.7, P = 0.003). First- or second-line immunotherapy patients have significantly longer PFS and OS than third- or fourth-line (PFS: 6.4 vs. 3.6, P = 0.009; OS: 20.0 vs. 10.5, P = 0.003). In patients with stage IV (n = 60) with extensive metastasis, the site of metastasis is mostly 1-3 sites after receiving toripalimab. The duration of PD-1 inhibitor OS in progressive patients (n = 56) was significantly prolonged (P = 0.038). CONCLUSION For patients with lung cancer, toripalimab can considerably extend PFS and OS in the first or second line and in stage III. PD-1 inhibitors are administered to patients with stage IV extensively metastatic lung cancer, which indicates an oligometastatic progression pattern, primarily in 1-3 locations, who are treated with PD-1 inhibitors. Continuing toripalimab beyond disease progression significantly prolonged OS.
Collapse
Affiliation(s)
- Chenlin Wang
- Department of Oncology, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Ning Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Lili Qiao
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Ya'nan Wu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Jiandong Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Yan Zhang
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
30
|
Logeart J, Samaille T, Falcoz A, Svrcek M, Dubreuil O, Vernerey D, Cohen R, Cervera P, Valverde A, Parc Y, André T. Survival Outcomes in Patients with Monobloc-Resected Stage IIC (pT4bN0) Colon Cancer: A Retrospective Observational Cohort Study. Clin Colorectal Cancer 2024; 23:346-353.e1. [PMID: 38853098 DOI: 10.1016/j.clcc.2024.05.005] [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: 12/12/2023] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND Stage II colon cancer (CC) exhibits considerable prognostic heterogeneous. Our objective was to assess survival but also the prognosis impact of microsatellite instability (MSI) in patients with stage IIC (T4bN0M0) CC. PATIENTS AND METHODS We conducted a retrospective observational study including all patients who had primary stage IIC CC resection between 2010 and 2020 in 2 expert centers. The primary endpoint was overall survival (OS) and disease-free survival (DFS) and time-to-relapse (TTR) were secondary endpoints. RESULTS Sixty-six patients, median age of 74 years [30-95], were included, with 37.9% presenting MSI (n = 25). Organ invasion involved the last ileal loop (n = 17), another colonic segment (n = 15), omentum (n = 13), visceral peritoneum (n = 13), and the bladder (n = 4). Surgical quality criteria showed complete monobloc resection in all patients and 93.9% R0 resection. After a median follow-up of 5 years [3.5-6.6], the entire population showed a 5-year OS of 65.2% [53.0-80.3] and 5-year DFS of 53.5% [41.1-69.6], with 18.9% [6.8-29.4] experiencing relapses at 5 years. The MSI phenotype correlated with improved 5-year OS (75.5% [56.5-100] vs. 59.5% [44.9-79.0], HR 0.41 [0.17-0.99]; P = .04), but DFS and TTR did not differ. Adjuvant chemotherapy was administered to 34.9% of patients. Univariate analysis identified age > 65 years, MSI status, and the number of nodes as factors associated with OS. CONCLUSION These data underline, in relation to a low rate of relapse, the lack of consensus regarding the appropriate indication for adjuvant chemotherapy in this high-risk stage II population.
Collapse
Affiliation(s)
- Juliette Logeart
- Sorbonne université, Departement of Medical Oncology, Saint Antoine Hospital, APHP, Paris, France; INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Sorbonne Université, Paris, France
| | - Thomas Samaille
- Sorbonne université, Departement of Medical Oncology, Saint Antoine Hospital, APHP, Paris, France
| | - Antoine Falcoz
- Department of Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France; University Bourgogne Franche-Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Magali Svrcek
- Department of Pathology, Saint-Antoine Hospital, AP-HP, Sorbonne Université, Paris, France
| | - Olivier Dubreuil
- Department of Digestive Oncology, Groupe Hospitalier Diaconesses Croix Saint Simon, Paris, France
| | - Dewi Vernerey
- Department of Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France; University Bourgogne Franche-Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Romain Cohen
- Sorbonne université, Departement of Medical Oncology, Saint Antoine Hospital, APHP, Paris, France; INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Sorbonne Université, Paris, France
| | - Pascale Cervera
- University Bourgogne Franche-Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Alain Valverde
- Department of Digestive Surgery, Groupe Hospitalier Diaconesses Croix Saint Simon, Paris, France
| | - Yann Parc
- Department of Digestive Surgery, AP-HP, Saint-Antoine Hospital, Sorbonne Université, Paris, France
| | - Thierry André
- Sorbonne université, Departement of Medical Oncology, Saint Antoine Hospital, APHP, Paris, France; INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Sorbonne Université, Paris, France.
| |
Collapse
|
31
|
Li Y, Tan L, Chen N, Liu X, Liang F, Yao Y, Zhang X, Wu A. Neoadjuvant Immunotherapy Alone for Patients With Locally Advanced and Resectable Metastatic Colorectal Cancer of dMMR/MSI-H Status. Dis Colon Rectum 2024; 67:1413-1422. [PMID: 39260435 DOI: 10.1097/dcr.0000000000003290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
BACKGROUND The use of programmed death-1 blockade has a significant therapeutic effect in patients with mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. However, data on preoperative single-agent programmed death-1 blockade are rare. OBJECTIVE This study aims to evaluate the effectiveness and safety of preoperative programmed death-1 blockade as a conversion strategy in patients with locally advanced and resectable metastatic mismatch repair-deficient/microsatellite instability-high colorectal cancer. DESIGN This is a retrospective observational study. SETTINGS This study was conducted at a high-volume tertiary referral cancer center in China. PATIENTS Twenty-four patients of consecutive cases since 2020 to 2022 with mismatch repair-deficient/microsatellite instability-high colorectal cancer who received preoperative single-agent programmed death-1 blockade were retrospectively reviewed. These patients had either bulking tumors scheduled for multivisceral resection, a strong desire for organ preservation, or potentially resectable metastatic lesions. MAIN OUTCOME MEASURES Pathological complete response, clinical complete response, toxicity, R0 resection rate, and complications were evaluated. RESULTS Patients tolerated preoperative immunotherapy well. The R0 resection rate was 95.2%, and the pathological complete response rate was 47.6%. Three patients (12.5%) were evaluated as having a clinical complete response and then underwent "watch and wait." One-half of the patients with cT4b were spared multivisceral resection, whereas 60% (3/5) achieved pathological complete response. All 3 patients with liver metastases obtained complete response of all liver lesions after programmed death-1 blockade treatment. Grade III postoperative complications occurred in 2 patients. LIMITATIONS The limitations of this study are as follows: retrospective study, small sample size, and short follow-up. CONCLUSIONS Preoperative anti-programmed death-1 therapy alone as a conversion strategy in initially resected difficult mismatch repair-deficient/microsatellite instability-high colorectal cancer can achieve a high tumor complete response. The use of immunopreoperative therapy in patients with T4b colon cancer or low rectal cancer can reduce multivisceral resection and achieve high organ function preservation. See the Video Abstract . INMUNOTERAPIA NEOADYUVANTE SOLA PARA PACIENTES CON CNCER COLORRECTAL LOCALMENTE AVANZADO Y METASTSICO RESECABLE CON ESTADO DMMR/MSIH ANTECEDENTES:El uso del bloqueo de muerte programada-1 tiene un efecto terapéutico significativo en pacientes con cáncer colorrectal metastásico deficiente en reparación de desajustes/inestabilidad de microsatélites-alta (dMMR/MSI-H). Sin embargo, los datos sobre el bloqueo preoperatorio de muerte programada-1 con un solo agente son escasos.OBJETIVO:Este estudio tiene como objetivo evaluar la eficacia y seguridad del bloqueo preoperatorio de muerte programada-1 como estrategia de conversión en pacientes con cáncer colorrectal localmente avanzado y metastásico resecable con dMMR/MSI-H.DISEÑO:Este es un estudio observacional retrospectivo.ESCENARIO:Este estudio se realizó en un centro oncológico terciario de referencia de gran volumen en China.PACIENTES:Se revisaron retrospectivamente veinticuatro pacientes de casos consecutivos desde 2020-2022 con cáncer colorrectal y dMMR/MSI-H que recibieron bloqueo preoperatorio de muerte programada-1 con un solo agente. Estos pacientes tenían un tumor voluminoso programado para resección multivisceral, un fuerte deseo de preservación del órgano o lesiones metastásicas potencialmente resecables.PRINCIPALES MEDIDAS DE RESULTADO:Se evaluaron la respuesta patológica completa, la respuesta clínica completa, la toxicidad, la tasa de resección R0 y las complicaciones.RESULTADOS:Los pacientes toleraron bien la inmunoterapia preoperatoria. La tasa de resección R0 fue del 95,2% y la tasa de respuesta patológica completa fue del 47,6%. Tres pacientes (12,5%) fueron evaluados como respuesta clínica completa y luego sometidos a "observar y esperar". La mitad de los pacientes cT4b se salvaron de la resección multivisceral, mientras que el 60% (3/5) lograron una respuesta patológica completa. Los tres pacientes con metástasis hepáticas obtuvieron respuesta completa de todas las lesiones hepáticas después del tratamiento de bloqueo de muerte programada-1. En dos pacientes se produjeron complicaciones postoperatorias de grado III.LIMITACIONES:Las limitaciones de este estudio son las siguientes: estudio retrospectivo, tamaño de muestra pequeño y seguimiento corto.CONCLUSIONES:La terapia preoperatoria anti muerte programada-1 sola como estrategia de conversión en el cáncer colorrectal inicialmente difícil de resecar con dMMR/MSI-H puede lograr una alta respuesta completa tumoral. El uso de terapia inmunopreoperatoria en pacientes con cáncer de colon T4b o cáncer de recto bajo puede reducir la resección multivisceral y lograr una alta preservación de la función del órgano. (Traducción-Dr. Felipe Bellolio ).
Collapse
Affiliation(s)
- Yingjie Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital and Institute, Beijing, China
| | - Luxin Tan
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Nan Chen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xinzhi Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital and Institute, Beijing, China
| | - Fei Liang
- Department of Biostatistics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunfeng Yao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaoyan Zhang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Aiwen Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
32
|
de Gooyer PGM, Verschoor YL, van den Dungen LDW, Balduzzi S, Marsman HA, Geukes Foppen MH, Grootscholten C, Dokter S, den Hartog AG, Verbeek WHM, Woensdregt K, van den Broek JJ, Oosterling SJ, Schumacher TN, Kuhlmann KFD, Beets-Tan RGH, Haanen JBAG, van Leerdam ME, van den Berg JG, Chalabi M. Neoadjuvant nivolumab and relatlimab in locally advanced MMR-deficient colon cancer: a phase 2 trial. Nat Med 2024; 30:3284-3290. [PMID: 39278994 PMCID: PMC11564102 DOI: 10.1038/s41591-024-03250-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 09/18/2024]
Abstract
Mismatch repair deficiency (dMMR) is found in approximately 15% of non-metastatic colon cancers (CCs) and is characterized by a defective DNA mismatch repair system, resulting in hypermutated and highly immunogenic tumors. Although patients with dMMR CC have limited benefit from chemotherapy, these tumors have been shown to respond exceptionally well to neoadjuvant anti-PD-1 plus anti-CTLA-4, with high rates of pathologic responses. Here, based on data from melanoma studies, we postulated a high efficacy and favorable toxicity profile of anti-PD-1 plus anti-LAG-3. In the NICHE-3 study, a total of 59 patients with locally advanced dMMR CC were treated with two 4-weekly cycles of nivolumab (480 mg) plus relatlimab (480 mg) before surgery. Pathologic response was observed in 57 of 59 (97%; 95% confidence interval (CI): 88-100%) patients, meeting the primary endpoint. Responses included 54 (92%; 95% CI: 81-97%) major pathologic responses (≤10% residual viable tumor) and 40 (68%; 95% CI: 54-79%) pathologic complete responses. With a median follow-up of 8 months (range, 2-19), one patient had recurrence of disease. The treatment displayed an acceptable safety profile, with all-grade and grade 3-4 immune-related adverse events (irAEs) occurring in 80% and 10% of patients, respectively. The most common irAEs were infusion-related reactions (29%), thyroid dysfunction (22%) and fatigue (20%). In conclusion, our results show that neoadjuvant nivolumab/relatlimab induces high rates of pathologic responses and that further investigation of this treatment in larger studies is warranted. These data add to the body of evidence in support of neoadjuvant immunotherapy regimens in dMMR CC. ClinicalTrials.gov identifier: NCT03026140 .
Collapse
Affiliation(s)
- Peter G M de Gooyer
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Yara L Verschoor
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Sara Balduzzi
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Marnix H Geukes Foppen
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Cecile Grootscholten
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Simone Dokter
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Anne G den Hartog
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wieke H M Verbeek
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Steven J Oosterling
- Department of Surgical Oncology, Spaarne Gasthuis, Haarlem and Hoofddorp, The Netherlands
| | - Ton N Schumacher
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Koert F D Kuhlmann
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Regina G H Beets-Tan
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
- Melanoma Clinic, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Monique E van Leerdam
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jose G van den Berg
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Myriam Chalabi
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Yang Z, Ma J, Han J, Li A, Liu G, Sun Y, Zheng J, Zhang J, Chen G, Xu R, Sun L, Meng C, Gao J, Bai Z, Deng W, Zhang C, Su J, Yao H, Zhang Z. Gut microbiome model predicts response to neoadjuvant immunotherapy plus chemoradiotherapy in rectal cancer. MED 2024; 5:1293-1306.e4. [PMID: 39047732 DOI: 10.1016/j.medj.2024.07.002] [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: 07/05/2023] [Revised: 02/18/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Accurate evaluation of the response to preoperative treatment enables the provision of a more appropriate personalized therapeutic schedule for locally advanced rectal cancer (LARC), which remains an enormous challenge, especially neoadjuvant immunotherapy plus chemoradiotherapy (nICRT). METHODS This prospective, multicenter cohort study enrolled patients with LARC from 6 centers who received nICRT. The dynamic variation in the gut microbiome during nICRT was evaluated. A species-level gut microbiome prediction (SPEED) model was developed and validated to predict the pathological complete response (pCR) to nICRT. FINDINGS A total of 50 patients were enrolled, 75 fecal samples were collected from 33 patients at different time points, and the pCR rate reached 42.4% (14/33). Lactobacillus and Eubacterium were observed to increase after nICRT. Additionally, significant differences in the gut microbiome were observed between responders and non-responders at baseline. Significantly higher abundances of Lachnospiraceae bacterium and Blautia wexlerae were found in responders, while Bacteroides, Prevotella, and Porphyromonas were found in non-responders. The SPEED model showcased a superior predictive performance with areas under the curve of 98.80% (95% confidence interval [CI]: 95.67%-100%) in the training cohort and 77.78% (95% CI: 65.42%-88.29%) in the validation cohort. CONCLUSIONS Programmed death 1 (PD-1) blockade plus concurrent long-course CRT showed a favorable pCR rate and is well tolerated in microsatellite-stable (MSS)/mismatch repair-proficient (pMMR) patients with LARC. The SPEED model can be used to predict the pCR to nICRT based on the baseline gut microbiome with high robustness and accuracy, thereby assisting clinical physicians in providing individualized management for patients with LARC. FUNDING This research was funded by the China National Natural Science Foundation (82202884).
Collapse
Affiliation(s)
- Zhengyang Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jingxin Ma
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jiagang Han
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ang Li
- Department of General Surgery, Beijing Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Gang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi Sun
- Department of Anorectal, Tianjin People's Hospital, Tianjin, China
| | - Jianyong Zheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jie Zhang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Guangyong Chen
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Rui Xu
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Liting Sun
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Cong Meng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jiale Gao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhigang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Wei Deng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Chenlin Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jianrong Su
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Hongwei Yao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China.
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China.
| |
Collapse
|
34
|
Hu H, Zhang J, Shi L, Huang M, Wang J, Deng Y. Reply to Y.-Y. Qiu et al. J Clin Oncol 2024; 42:3510-3511. [PMID: 39047220 DOI: 10.1200/jco-24-01217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 07/27/2024] Open
Affiliation(s)
- Huabin Hu
- Huabin Hu, MD and Jianwei Zhang, MD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Lishuo Shi, PhD, Clinical Research Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Meijin Huang, MD and Jianping Wang, MD, 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; and Yanhong Deng, MD, PhD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jianwei Zhang
- Huabin Hu, MD and Jianwei Zhang, MD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Lishuo Shi, PhD, Clinical Research Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Meijin Huang, MD and Jianping Wang, MD, 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; and Yanhong Deng, MD, PhD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Lishuo Shi
- Huabin Hu, MD and Jianwei Zhang, MD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Lishuo Shi, PhD, Clinical Research Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Meijin Huang, MD and Jianping Wang, MD, 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; and Yanhong Deng, MD, PhD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Meijin Huang
- Huabin Hu, MD and Jianwei Zhang, MD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Lishuo Shi, PhD, Clinical Research Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Meijin Huang, MD and Jianping Wang, MD, 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; and Yanhong Deng, MD, PhD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jianping Wang
- Huabin Hu, MD and Jianwei Zhang, MD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Lishuo Shi, PhD, Clinical Research Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Meijin Huang, MD and Jianping Wang, MD, 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; and Yanhong Deng, MD, PhD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanhong Deng
- Huabin Hu, MD and Jianwei Zhang, MD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Lishuo Shi, PhD, Clinical Research Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Meijin Huang, MD and Jianping Wang, MD, 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; and Yanhong Deng, MD, PhD, 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, and Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
35
|
Huang W, Zhang S. A watch-and-wait approach for metachronous multiple colon cancer following neoadjuvant immunotherapy: a case report. Front Immunol 2024; 15:1391038. [PMID: 39430753 PMCID: PMC11486642 DOI: 10.3389/fimmu.2024.1391038] [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: 02/24/2024] [Accepted: 09/06/2024] [Indexed: 10/22/2024] Open
Abstract
The application of immunotherapy for treating colorectal cancer (CRC) is currently a research hotspot, and neoadjuvant immunotherapy has shown initial success in treating CRC. The watch-and-wait (W&W) approach is often used after achieving a clinical complete response (cCR) following preoperative treatment of low rectal cancer. However, thus far, the W&W approach has not been reported for patients with colon cancer. Here, we report the case of a 64-year-old patient with heterogeneous multigenic CRC who achieved cCR after five sessions of neoadjuvant immunotherapy before surgery. A W&W approach was used to spare the patient from surgery. A 64-year-old male presented with intermittent abdominal pain. A colonoscopy examination detected an irregular cauliflower-like mass near the hepatic flexure of the ascending colon. The biopsy results indicated adenocarcinoma of the ascending colon. The patient was administered pembrolizumab (200 mg, ivgtt, q3w). After one cycle of treatment, the intestinal obstruction symptoms disappeared, and the treatment was continued for additional three sessions. After complete clinical remission of the tumor was confirmed, the W&W approach was adopted. Follow-up CT scans and colonoscopy examinations confirmed no local tumor regeneration or metastasis. Neoadjuvant immunotherapy is effective for patients with DNA mismatch repair gene deficiency and/or microsatellite instability high with a high rate of cCR or pathologic complete response. The W&W approach may also be suitable for patients with colon cancer. The safety and feasibility of watch and wait in patients with colon cancer need to be verified by more clinical data.
Collapse
Affiliation(s)
| | - Shouru Zhang
- Department of Gastrointestinal Surgery, Chongqing University Cancer Hospital, Chongqing, ;China
| |
Collapse
|
36
|
Xia F, Wang Y, Wang H, Shen L, Xiang Z, Zhao Y, Zhang H, Wan J, Zhang H, Wang Y, Wu R, Wang J, Yang W, Zhou M, Zhou S, Chen Y, Zhang Z, Wu X, Xuan Y, Wang R, Sun Y, Tong T, Zhang X, Wang L, Huang D, Sheng W, Yan H, Yang X, Shen Y, Xu Y, Zhao R, Mo M, Cai G, Cai S, Xu Y, Zhang Z. Randomized Phase II Trial of Immunotherapy-Based Total Neoadjuvant Therapy for Proficient Mismatch Repair or Microsatellite Stable Locally Advanced Rectal Cancer (TORCH). J Clin Oncol 2024; 42:3308-3318. [PMID: 38950321 DOI: 10.1200/jco.23.02261] [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: 10/17/2023] [Revised: 01/17/2024] [Accepted: 04/17/2024] [Indexed: 07/03/2024] Open
Abstract
PURPOSETo assess whether the integration of PD-1 inhibitor with total neoadjuvant therapy (iTNT) can lead to an improvement in complete responses (CRs) and favors a watch-and-wait (WW) strategy in patients with proficient mismatch repair or microsatellite stable (pMMR/MSS) locally advanced rectal cancer (LARC).PATIENTS AND METHODSWe conducted a prospective, multicenter, randomized, open-label, phase II trial using a pick-the-winner design. Eligible patients with clinical T3-4 and/or N+ rectal adenocarcinoma were randomly assigned to group A for short-course radiotherapy (SCRT) followed by six cycles of consolidation immunochemotherapy with capecitabine and oxaliplatin and toripalimab or to group B for two cycles of induction immunochemotherapy followed by SCRT and the rest four doses. Either total mesorectal excision or WW was applied on the basis of tumor response. The primary end point was CR which included pathological CR (pCR) after surgery and clinical CR (cCR) if WW was applicable, with hypothesis of an increased CR of 40% after iTNT compared with historical data of 25% after conventional TNT.RESULTSOf the 130 patients enrolled, 121 pMMR/MSS patients were evaluable (62 in group A and 59 in group B). At a median follow-up of 19 months, CR was achieved at 56.5% in group A and 54.2% in group B. Both groups fulfilled the predefined statistical hypothesis (P < .001). Both groups reported a pCR rate of 50%. Respectively, 15 patients in each group underwent WW and remained disease free. The most frequent grade 3 to 4 toxicities were thrombocytopenia and neutropenia. Patients in group A had higher rate of cCR (43.5% v 35.6%) at restaging and lower rate of grade 3 to 4 thrombocytopenia (24.2% v 33.9%) during neoadjuvant treatment.CONCLUSIONThe iTNT regimens remarkably improved CR rates in pMMR/MSS LARC compared with historical benchmark with acceptable toxicity. Up-front SCRT followed by immunochemotherapy was selected for future definitive study.
Collapse
Affiliation(s)
- Fan Xia
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yaqi Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Wang
- Department of Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Lijun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zuolin Xiang
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yutian Zhao
- Department of Radiation Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Huojun Zhang
- Department of Radiation Oncology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Juefeng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruiyan Wu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingwen Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wang Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Menglong Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shujuan Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yajie Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xian Wu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Xuan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Renjie Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yiqun Sun
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Tong Tong
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xun Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lei Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Dan Huang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Weiqi Sheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Hao Yan
- Department of Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Xu Yang
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuxin Shen
- Department of Radiation Oncology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yu Xu
- Department of Radiation Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ruping Zhao
- Department of Radiation Oncology, Jia Hui International Hospital, Shanghai, China
- Department of Radiotherapy, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Miao Mo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Cancer Prevention, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Guoxiang Cai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Sanjun Cai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ye Xu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Roxburgh CS. Time to Change Strategy in Non-metastatic Locally Advanced Mismatch Repair-Deficient Colon Cancer. Ann Surg Oncol 2024; 31:6342-6343. [PMID: 38992311 DOI: 10.1245/s10434-024-15780-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024]
Affiliation(s)
- Campbell S Roxburgh
- Colorectal Surgical Oncology, Glasgow Royal Infirmary, School of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
38
|
Deng Z, Luo Y, Chen X, Pan T, Rui Y, Hu H, Yan J, Zhang K, Luo C, Song B. Pathological response following neoadjuvant immunotherapy and imaging characteristics in dMMR/MSI-H locally advanced colorectal cancer. Front Immunol 2024; 15:1466497. [PMID: 39399495 PMCID: PMC11466765 DOI: 10.3389/fimmu.2024.1466497] [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: 07/18/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Background In recent years, there has been significant research interest in immunotherapy for colorectal cancer (CRC). Specifically, immunotherapy has emerged as the primary treatment for patients with mismatch repair gene defects (dMMR) or microsatellite highly unstable (MSI-H) who have colorectal cancer. Yet, there is currently no data to support the practicality and safety of neoadjuvant immunotherapy for colorectal cancer with dMMR or MSI-H. Therefore, a study was conducted to identify the postoperative pathology, safety profile, and imaging features of patients with dMMR or MSI-H CRC following neoadjuvant immunotherapy. Methods The retrospective study was carried out on patients with locally advanced or metastatic CRC who received immunotherapy at Sichuan Cancer Hospital, with approval from the hospital's ethics committee. The study aimed to assess the short-term effectiveness of immunotherapy by focusing on pathological complete response (pCR) as the primary outcome, while also considering secondary endpoints such as objective response rate, disease-free survival, and safety profile. Results Twenty patients with dMMR/MSI-H CRC who underwent neoadjuvant immunotherapy as part of the treatment were enrolled between May 2019 and February 2024 at Sichuan Cancer Hospital. Out of these patients, eight patients received PD-1 blockade monotherapy as neoadjuvant treatment, while 12 were administered a combined therapy of anti-CTLA-4 and anti-PD-1. 12 patients received Nivolumab plus Ipilimumab regimen and 8 patients received PD-1 blockades (2 patients were Pembrolizumab, 2 patients were Sintilimab, 4 patients were Tislelizumab) monotherapy. Additionally, 19 patients underwent surgery after immunotherapy and of these, 15 (75.0%) achieved complete pathological response (pCR), 8 (66.7%) achieved the same on Nivolumab plus Ipilimumab immunotherapy while 7 (87.5%) achieved on PD-1 antibody monotherapy. The overall response rate (ORR) was 75%, with 45.0% of patients experiencing grade I/II immunotherapy-related adverse events. The most frequent adverse event observed was increased ALT i.e. 20%. Notably, no postoperative complications were observed. Conclusion Based on the findings, neoadjuvant immunotherapy for colorectal cancer may be both safe and effective in clinical practice. Furthermore, the study suggested that dual immunotherapy could potentially increase the immunotherapy cycle and contribute to a superior pCR rate. However, the conclusion emphasized the need for further prospective clinical trials to validate these results.
Collapse
Affiliation(s)
- Zijian Deng
- Department of Colorectal Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, China
| | - Yajun Luo
- Department of Colorectal Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoli Chen
- Department of Imaging Department, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Pan
- Department of Colorectal Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, China
| | - Yuanyi Rui
- Department of Colorectal Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, China
| | - Hai Hu
- Department of Colorectal Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, China
| | - Jin Yan
- Department of Colorectal Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, China
| | - Ke Zhang
- Department of Colorectal Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, China
| | - Cheng Luo
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Bo Song
- Department of Colorectal Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
39
|
Xiao WW, Chen G, Gao YH, Lin JZ, Wu XJ, Luo HL, Lu ZH, Wang QX, Sun R, Cai PQ, Zhu CM, Liu M, Li JB, Wang YR, Jin Y, Wang F, Luo HT, Li CL, Pan ZZ, Xu RH. Effect of neoadjuvant chemoradiotherapy with or without PD-1 antibody sintilimab in pMMR locally advanced rectal cancer: A randomized clinical trial. Cancer Cell 2024; 42:1570-1581.e4. [PMID: 39094560 DOI: 10.1016/j.ccell.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 05/26/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024]
Abstract
Neoadjuvant chemoradiotherapy (NACRT) was the standard treatment for patients with locally advanced rectal cancer (LARC) with proficient mismatch repair (pMMR) proteins. In this randomized phase 2 trial (ClinicalTrial.gov: NCT04304209), 134 pMMR LARC patients were randomly (1:1) assigned to receive NACRT or NACRT and the programmed cell death protein 1 (PD-1) antibody sintilimab. As the primary endpoint, the total complete response (CR) rate is 26.9% (18/67, 95% confidence interval [CI] 16.0%-37.8%) and 44.8% (30/67, 95% CI 32.6%-57.0%) in the control and experimental arm, respectively, with significant difference (p = 0.031 for chi-squared test). Response ratio is 1.667 (95% CI 1.035-2.683). Immunohistochemistry shows PD-1 ligand 1 (PD-L1) combined positive score is associated with the synergistic effect. The safety profile is similar between the arms. Adding the PD-1 antibody sintilimab to NACRT significantly increases the CR rate in pMMR LARC, with a manageable safety profile. PD-L1 positivity may help identify patients who might benefit most from the combination therapy.
Collapse
Affiliation(s)
- Wei-Wei Xiao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China; United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co., Ltd, Guangzhou, China
| | - Gong Chen
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yuan-Hong Gao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Jun-Zhong Lin
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xiao-Jun Wu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Hui-Long Luo
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Zhen-Hai Lu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Qiao-Xuan Wang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Rui Sun
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Pei-Qiang Cai
- Department of Radiology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Chong-Mei Zhu
- Department of Pathology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Min Liu
- Department of Ultrasound, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ji-Bin Li
- Department of Statistics, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yi-Rui Wang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ying Jin
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Feng Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Hai-Tao Luo
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, China
| | - Cai-Ling Li
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, China
| | - Zhi-Zhong Pan
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
40
|
Platt JR, Allotey J, Alouani E, Glasbey J, Intini R, Lonardi S, Mazzoli G, Militello AM, Modest DP, Palle J, Pietrantonio F, Riyad K, Samuel L, Schulze AV, Shiu KK, Taieb J, Tolan DJM, West NP, Westwood AC, Williams CJM, Seligmann JF. Risk of bowel obstruction in patients with colon cancer responding to immunotherapy: an international case series. ESMO Open 2024; 9:103698. [PMID: 39241497 PMCID: PMC11408065 DOI: 10.1016/j.esmoop.2024.103698] [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: 06/29/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Immunotherapy is used routinely for treating deficient mismatch repair (dMMR) colon cancer (CC). This case series highlights an emerging safety issue, where patients develop bowel obstruction associated with immunotherapy response. PATIENTS AND METHODS Patients with dMMR CC who developed bowel obstruction while responding to immunotherapy were retrospectively identified. Data on patient, disease, treatment, and response-specific factors were explored for potential risk factors. Overall treatment numbers were used to estimate incidence. RESULTS Nine patients from eight European centres were included. Common features were hepatic flexure location (5/9), T4 radiological staging (6/9), annular shape (8/9), radiological stricturing (5/9), and endoscopic obstruction (6/9). All received pembrolizumab and obstructed between 45 and 652 days after starting treatment. Seven patients underwent surgical resection; one was managed with a defunctioning stoma; and one was managed conservatively. One patient died from obstruction. Radiological response was seen in eight patients, including two complete responses. Pathological response was seen in all seven who underwent resection, including four complete responses. The overall incidence of immunotherapy response-related obstruction in these centres was 1.51%. CONCLUSIONS Bowel obstruction associated with immunotherapy response may represent a rare treatment-related complication in dMMR CC. Clinicians must recognise this safety signal and share experience to maintain patient safety.
Collapse
Affiliation(s)
- J R Platt
- Division of Oncology, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds.
| | - J Allotey
- Department of Oncology, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, UK
| | - E Alouani
- Digestive Oncology Department, Rangueil Hospital, University Hospital of Toulouse, Toulouse, France
| | - J Glasbey
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK. https://twitter.com/DrJamesGlasbey
| | - R Intini
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua
| | - S Lonardi
- Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua
| | - G Mazzoli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - A M Militello
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - D P Modest
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin; German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - J Palle
- Université Paris Cité, Digestive Oncology Department, Hôpital Européen Georges Pompidou, Paris; Université Paris Cité, Pancreatology and Digestive Oncology Department, Hôpital Beaujon, Clichy, France
| | - F Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - K Riyad
- The John Goligher Colorectal Surgery Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - L Samuel
- Department of Oncology, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, UK
| | - A V Schulze
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin
| | - K K Shiu
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - J Taieb
- Institut du Cancer Paris CARPEM, Gastroenterology and Digestive Oncology Department, APHP Centre - Université Paris Cité, Hôpital Européen Georges Pompidou, Paris, France
| | - D J M Tolan
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds
| | - N P West
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - A C Westwood
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - C J M Williams
- Division of Oncology, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds. https://twitter.com/chrisjmwilliams
| | - J F Seligmann
- Division of Oncology, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds
| |
Collapse
|
41
|
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
|
42
|
Aktar T, Modak S, Majumder D, Maiti D. A detailed insight into macrophages' role in shaping lung carcinogenesis. Life Sci 2024; 352:122896. [PMID: 38972632 DOI: 10.1016/j.lfs.2024.122896] [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: 03/05/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
Despite significant advancements in cancer treatment in recent decades, the high mortality rate associated with lung cancer remains a significant concern. The development and proper execution of new targeted therapies needs more deep knowledge regarding the lung cancer associated tumour microenvironment. One of the key component of that tumour microenvironment is the lung resident macrophages. Although in normal physiological condition the lung resident macrophages are believed to maintain lung homeostasis, but they may also initiate a vicious inflammatory response in abnormal conditions which is linked to lung cancer development. Depending on the activation pathway, the lung resident macrophages are either of M1 or M2 sub-type. The M1 and M2 sub-types differ significantly in various prospectuses, from phenotypic markers to metabolic pathways. In addition to this generalized classification, the recent advancement of the multiomics technology is able to identify some other sub-types of lung resident macrophages. Researchers have also observed that these different sub-types can manipulate the pathogenesis of lung carcinogenesis in a context dependent manner and can either promote or inhibit the development of lung carcinogenesis upon receiving proper activation. As proper knowledge about the role played by the lung resident macrophages' in shaping the lung carcinogenesis is limited, so the main purpose of this review is to bring all the available information under the same roof. We also elaborated the different mechanisms involved in maintenance of the plasticity of M1/M2 sub-type, as this plasticity can be a good target for lung cancer treatment.
Collapse
Affiliation(s)
- Tamanna Aktar
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Snehashish Modak
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Debabrata Majumder
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India; Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Debasish Maiti
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India.
| |
Collapse
|
43
|
Li J. Clinical status and future prospects of neoadjuvant immunotherapy for localized mismatch repair-deficient cancers: a review. Int J Surg 2024; 110:5722-5732. [PMID: 38768473 PMCID: PMC11392202 DOI: 10.1097/js9.0000000000001680] [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: 03/27/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Frameshift mutations accumulate in cancers related to mismatch repair deficiency (dMMR), which has the potential to produce various neoantigens, representing a distinct subset of cancers that respond considerably to immunotherapy. In recent years, robust evidence has supported the first-line application of immunotherapy for patients with metastatic dMMR cancers, which provoked extensive investigations of the feasibility and efficacy of immunotherapy in up-front settings, including neoadjuvant therapy. Several completed trials with small sample sizes suggested that neoadjuvant immunotherapy can achieve an impressively high complete response rate, for the first time offering the potential of systemic therapy to cure cancer without the need for surgical resection. However, a difficult dilemma emerges: clinicians are now facing a selection between the standard of care with good evidence for proficient MMR but suboptimal for dMMR cancers and the emerging immunotherapy with promising results but only based on a limited number of patients with shorter duration of follow-up. This review aims to provide a comprehensive summary of the biological rationale and clinical status of neoadjuvant immunotherapy in patients with dMMR cancers. Furthermore, I elaborate on particular issues that must be taken into consideration for further advancement in the field.
Collapse
Affiliation(s)
- Jian Li
- Department of General Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan, People’s Republic of China
| |
Collapse
|
44
|
Fan WX, Su F, Zhang Y, Zhang XL, Du YY, Gao YJ, Li WL, Hu WQ, Zhao J. Oncological characteristics, treatments and prognostic outcomes in MMR-deficient colorectal cancer. Biomark Res 2024; 12:89. [PMID: 39183366 PMCID: PMC11346251 DOI: 10.1186/s40364-024-00640-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent cancer globally. It's recognized that the molecular subtype of CRC, characterized by mismatch repair deficiency (dMMR) or microsatellite instability-high (MSI-H), plays a critical role in determining appropriate treatment strategies. This review examines the current molecular classifications, focusing on dMMR/MSI-H CRC and its subtypes: Lynch syndrome (LS), Lynch-like syndrome (LLS), and sporadic cases. Despite advances in understanding of these genetic backgrounds, clinical trials have not conclusively differentiated the efficacy of immune checkpoint inhibitors among these subgroups. Therefore, while this review details the molecular characteristics and their general implications for treatment and prognosis, it also highlights the limitations and the need for more refined clinical studies to ascertain tailored therapeutic strategies for each subtype. Furthermore, this review summarizes completed and ongoing clinical studies, emphasizing the importance of developing treatments aligned more closely with molecular profiles. By discussing these aspects, the review seeks to provide a comprehensive analysis of oncological characteristics, presenting a detailed understanding of their implications for treatment and prognosis in dMMR/MSI-H CRC.
Collapse
Affiliation(s)
- Wen-Xuan Fan
- Graduate School of Shanxi Medical University, Taiyuan, Shanxi, 030607, China
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Fei Su
- Graduate School of Shanxi Medical University, Taiyuan, Shanxi, 030607, China
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Yan Zhang
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
- Graduate School of Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Xiao-Ling Zhang
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Yun-Yi Du
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Yang-Jun Gao
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Wei-Ling Li
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
- Graduate School of Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Wen-Qing Hu
- Department of Gastrointestinal Surgery, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Jun Zhao
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China.
| |
Collapse
|
45
|
Chong X, Madeti Y, Cai J, Li W, Cong L, Lu J, Mo L, Liu H, He S, Yu C, Zhou Z, Wang B, Cao Y, Wang Z, Shen L, Wang Y, Zhang X. Recent developments in immunotherapy for gastrointestinal tract cancers. J Hematol Oncol 2024; 17:65. [PMID: 39123202 PMCID: PMC11316403 DOI: 10.1186/s13045-024-01578-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
The past few decades have witnessed the rise of immunotherapy for Gastrointestinal (GI) tract cancers. The role of immune checkpoint inhibitors (ICIs), particularly programmed death protein 1 (PD-1) and PD ligand-1 antibodies, has become increasingly pivotal in the treatment of advanced and perioperative GI tract cancers. Currently, anti-PD-1 plus chemotherapy is considered as first-line regimen for unselected advanced gastric/gastroesophageal junction adenocarcinoma (G/GEJC), mismatch repair deficient (dMMR)/microsatellite instability-high (MSI-H) colorectal cancer (CRC), and advanced esophageal cancer (EC). In addition, the encouraging performance of claudin18.2-redirected chimeric antigen receptor T-cell (CAR-T) therapy in later-line GI tract cancers brings new hope for cell therapy in solid tumour treatment. Nevertheless, immunotherapy for GI tumour remains yet precise, and researchers are dedicated to further maximising and optimising the efficacy. This review summarises the important research, latest progress, and future directions of immunotherapy for GI tract cancers including EC, G/GEJC, and CRC.
Collapse
Affiliation(s)
- Xiaoyi Chong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yelizhati Madeti
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jieyuan Cai
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Wenfei Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Cong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jialin Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Liyang Mo
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Huizhen Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Siyi He
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Chao Yu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhiruo Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Boya Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yakun Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
46
|
Wu Z, Zhang Y, Cheng Y, Li J, Li F, Wang C, Shi L, Qin G, Zhan W, Cai Y, Xie X, Ling J, Hu H, Zhang J, Deng Y. PD-1 blockade plus COX inhibitors in dMMR metastatic colorectal cancer: Clinical, genomic, and immunologic analyses from the PCOX trial. MED 2024; 5:998-1015.e6. [PMID: 38795703 DOI: 10.1016/j.medj.2024.05.002] [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: 12/15/2023] [Revised: 04/08/2024] [Accepted: 05/02/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Approximately 20% of patients with DNA mismatch repair deficiency (dMMR) metastatic colorectal cancer do not respond to anti-programmed death-1 (PD-1) ligand therapy, and baseline biomarkers of response are lacking. METHODS We conducted a phase 2 study to evaluate the efficacy of cyclooxygenase (COX) inhibitors in combination with anti-PD-1 therapy in patients with dMMR metastatic colorectal cancer. The primary endpoint was objective response rate. The secondary endpoints included progression-free survival (PFS), overall survival (OS), disease control rate, duration of response, and safety. FINDINGS A total of 30 patients were enrolled, and the objective response rate was 73.3%, meeting the predefined endpoint of 68%. The median PFS and median OS were not reached at a median follow-up period of 50.8 months. Disease control was achieved in 28 patients (93.3%). The median duration of response was not reached. The combination was well tolerated. Multiomics analysis revealed that the antigen processing and presentation pathway was positively associated with treatment response and PFS. Higher TAPBP expression was predictive of better PFS (log-rank p = 0.003), and this prognostic significance was confirmed in an immunotherapy validation cohort. CONCLUSIONS Thus, COX inhibitors combined with PD-1 blockade may be effective and safe treatment options for patients with dMMR metastatic colorectal cancer, and TAPBP may serve as a biomarker for immune checkpoint inhibitor therapy (this study was registered at ClinicalTrials.gov: NCT03638297). FUNDING Funded by the National Natural Science Foundation of China (81974369) and the program of Guangdong Provincial Clinical Research Center for Digestive Diseases (2020B1111170004).
Collapse
Affiliation(s)
- Zehua Wu
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yuanzhe Zhang
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yi Cheng
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Jianxia Li
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Fangqian Li
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Chao Wang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Lishuo Shi
- Clinical Research Centre, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Ge Qin
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Weixiang Zhan
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yue Cai
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Xiaoyu Xie
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Jiayu Ling
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Huabin Hu
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Jianwei Zhang
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yanhong Deng
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; State Key Laboratory of Oncology in South China, Guangzhou 510060, China.
| |
Collapse
|
47
|
Duval M, Vanderbecq Q, Phou V, Cervantes B, Mas L, Bachet JB, Goumard C, Parc Y, André T, Lefèvre JH, Lucidarme O, Arrivé L, Cohen R, Wagner M. Performances of preoperative CT scan to predict the pTN stage for MSI/dMMR localized colon cancers. ESMO Open 2024; 9:103678. [PMID: 39146669 PMCID: PMC11374966 DOI: 10.1016/j.esmoop.2024.103678] [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: 05/28/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Neoadjuvant immunotherapy emerges as a promising strategy for patients with localized colon cancer (CC) harboring microsatellite instability/mismatch repair deficiency (MSI/dMMR). The aim of this study is to evaluate the concordance between clinical cTN stage assessed by preoperative computed tomography (CT) scan and pTN stage of MSI/dMMR CC. PATIENTS AND METHODS Consecutive patients diagnosed for localized MSI/dMMR CC and treated with upfront surgery between 2013 and 2022 in two French centers were eligible. Two independent radiologists, blinded to pathological findings, reviewed all preoperative CT scans and assessed cTN stage, with a third radiologist reviewing discordant cases. Radiological predictive diagnostic accuracy for pT4 and pN+ (N+ = N1 or N2) were calculated. RESULTS One hundred and thirteen patients were included (right CCs = 79%). CT scan diagnostic performances for pT4 were sensitivity (Se) = 33.3%; specificity (Sp) = 94.0%; positive predictive value (PPV) = 66.7%; and negative predictive value (NPV) = 79.6% and for pN+ were Se = 70.3%; Sp = 59.2%; PPV = 45.6%; and NPV = 80.4%. When pT-pN were combined, 37.5% of tumors identified as cT4 and/or cN+ were actually pT1-3 and pN0, and 23.1% of the pT4 and pN+ population was not identified as such radiologically. CONCLUSION The ability of preoperative CT scan to predict pT and pN stages is limited for localized MSI/dMMR CCs. Reassessing neoadjuvant strategies' benefit-risk balance in this population is needed.
Collapse
Affiliation(s)
- M Duval
- Sorbonne Université, Service d'Oncologie Médicale, Hôpital Saint Antoine, AP-HP, INSERM UMRS 938, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, SIRIC CURAMUS, Paris.
| | - Q Vanderbecq
- Sorbonne Université, Service de Radiologie, Hôpital Saint Antoine, APHP, Laboratoire d'Imagerie Biomédicale, UMR 7371, UMR S 1146, Paris
| | - V Phou
- Sorbonne Université, Service d'Imagerie (SISU), Hôpital Pitié Salpêtrière, APHP, Laboratoire d'Imagerie Biomédicale, UMR 7371, UMR_S 1146, Paris
| | - B Cervantes
- Sorbonne Université, Service d'Oncologie Médicale, Hôpital Saint Antoine, AP-HP, INSERM UMRS 938, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, SIRIC CURAMUS, Paris
| | - L Mas
- Sorbonne Université, Service d'Hépato-gastroentérologie et Oncologie Digestive, Hôpital Pitié Salpêtrière, APHP, Paris
| | - J-B Bachet
- Sorbonne Université, Service d'Hépato-gastroentérologie et Oncologie Digestive, Hôpital Pitié Salpêtrière, APHP, Paris
| | - C Goumard
- Sorbonne Université, Service de Chirurgie Digestive, Hépatobiliaire et Transplantation Hépatique, Hôpital Pitié Salpêtrière, Centre de Recherche Saint-Antoine, INSERM UMR S-938, Paris
| | - Y Parc
- Sorbonne Université, Service de Chirurgie Générale et Digestive, Hôpital Saint-Antoine, AP-HP, INSERM UMRS 938, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, SIRIC CURAMUS, Paris, France
| | - T André
- Sorbonne Université, Service d'Oncologie Médicale, Hôpital Saint Antoine, AP-HP, INSERM UMRS 938, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, SIRIC CURAMUS, Paris
| | - J H Lefèvre
- Sorbonne Université, Service de Chirurgie Générale et Digestive, Hôpital Saint-Antoine, AP-HP, INSERM UMRS 938, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, SIRIC CURAMUS, Paris, France
| | - O Lucidarme
- Sorbonne Université, Service d'Imagerie (SISU), Hôpital Pitié Salpêtrière, APHP, Laboratoire d'Imagerie Biomédicale, UMR 7371, UMR_S 1146, Paris
| | - L Arrivé
- Sorbonne Université, Service de Radiologie, Hôpital Saint Antoine, APHP, Laboratoire d'Imagerie Biomédicale, UMR 7371, UMR S 1146, Paris
| | - R Cohen
- Sorbonne Université, Service d'Oncologie Médicale, Hôpital Saint Antoine, AP-HP, INSERM UMRS 938, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, SIRIC CURAMUS, Paris
| | - M Wagner
- Sorbonne Université, Service d'Imagerie (SISU), Hôpital Pitié Salpêtrière, APHP, Laboratoire d'Imagerie Biomédicale, UMR 7371, UMR_S 1146, Paris
| |
Collapse
|
48
|
Sandow L, Tsikitis L, Lopez CD, Brinkerhoff B, Kardosh A, Pegna G, Chen EY. Neoadjuvant immunotherapy leads to complete pathologic response in locally advanced colon cancer. Clin Case Rep 2024; 12:e9218. [PMID: 39114842 PMCID: PMC11303659 DOI: 10.1002/ccr3.9218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/29/2024] [Accepted: 05/14/2024] [Indexed: 08/10/2024] Open
Abstract
Immunotherapy is considered first line in patients with dMMR metastatic colorectal cancer (CRC). Recent studies have also shown promising results with neoadjuvant immunotherapy in locally advanced CRC. We report a case in which neoadjuvant immunotherapy with pembrolizumab resulted in complete pathologic response at time of resection as well as saved the patient the morbidity associated with a hepatectomy. We also completed a scoping review of the literature which suggests promising tumor responses with treatment in dMMR CRC. Further randomized control trials to determine the magnitude of response and optimal regimen are needed.
Collapse
Affiliation(s)
- Lyndsey Sandow
- Department of MedicineOregon Health and Science UniversityPortlandOregonUSA
| | - Liana Tsikitis
- Division of Gastrointestinal and General Surgery, Department of General SurgeryOregon Health and Science UniversityPortlandOregonUSA
| | - Charles D. Lopez
- Division of Hematology and Medical OncologyOregon Health and Science University, Knight Cancer InstitutePortlandOregonUSA
| | - Brian Brinkerhoff
- Department of PathologyOregon Health and Science UniversityPortlandOregonUSA
| | - Adel Kardosh
- Division of Hematology and Medical OncologyOregon Health and Science University, Knight Cancer InstitutePortlandOregonUSA
| | - Guillaume Pegna
- Division of Hematology and Medical OncologyOregon Health and Science University, Knight Cancer InstitutePortlandOregonUSA
| | - Emerson Y. Chen
- Division of Hematology and Medical OncologyOregon Health and Science University, Knight Cancer InstitutePortlandOregonUSA
| |
Collapse
|
49
|
Zhu D, Feng H, Zhang Z, Li J, Li Y, Hou T. DEPDC1B: A novel tumor suppressor gene associated with immune infiltration in colon adenocarcinoma. Cancer Med 2024; 13:e70043. [PMID: 39087856 PMCID: PMC11292854 DOI: 10.1002/cam4.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Recent research indicates a positive correlation between DEP structural domain-containing 1B (DEPDC1B) and the cell cycle in various tumors. However, the role of DEPDC1B in the infiltration of the tumor immune microenvironment (TIME) remains unexplored. METHODS We analyzed the differential expression and prognostic significance of DEPDC1B in colon adenocarcinoma (COAD) using the R package "limma" and the Gene Expression Profiling Interactive Analysis (GEPIA) website. Gene set enrichment analysis (GSEA) was employed to investigate the functions and interactions of DEPDC1B expression in COAD. Cell Counting Kit-8 (CCK-8) assays and colony formation assays were utilized to assess the proliferative function of DEPDC1B. Correlations between DEPDC1B expression and tumor-infiltrating immune cells, immune checkpoints, tumor mutational burden (TMB), and microsatellite instability (MSI) status were examined using Spearman correlation analysis and CIBERSORT. RESULTS DEPDC1B was highly expressed in COAD. Elevated DEPDC1B expression was associated with lower epithelial-to-mesenchymal transition (EMT) and TNM stages, leading to a favorable prognosis. DEPDC1B mRNA was prominently expressed in COAD cell lines. CCK-8 and colony formation assays demonstrated that DEPDC1B inhibited the proliferation of COAD cells. Analysis using the CIBERSORT database and Spearman correlation revealed that DEPDC1B correlated with four types of tumor-infiltrating immune cells. Furthermore, high DEPDC1B expression was linked to the expression of PD-L1, CTLA4, SIGLEC15, PD-L2, TMB, and MSI-H. High DEPDC1B expression also indicated responsiveness to anti-PD-L1 immunotherapy. CONCLUSIONS DEPDC1B inhibits the proliferation of COAD cells and positively regulates the cell cycle, showing a positive correlation with CCNB1 and PBK expression. DEPDC1B expression in COAD is associated with tumor-infiltrating immune cells, immune checkpoints, TMB, and MSI-H in the tumor immune microenvironment. This suggests that DEPDC1B may serve as a novel prognostic marker and a potential target for immunotherapy in COAD.
Collapse
Affiliation(s)
- Dandan Zhu
- Guangdong Center for Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdongChina
| | - Huolun Feng
- School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdongChina
| | - Zhixiong Zhang
- Guangdong Center for Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdongChina
| | - Jiaqi Li
- Guangdong Center for Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdongChina
| | - Yong Li
- School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdongChina
| | - Tieying Hou
- Medical Experimental CenterShenzhen Nanshan People's HospitalShenzhenGuangdongChina
- Medical SchoolShenzhen UniversityShenzhenGuangdongChina
| |
Collapse
|
50
|
Gu W, Zeng D, Zhang C. Discovering the effect of combination of celecoxib and sorafenib on hepatocellular carcinoma. Discov Oncol 2024; 15:321. [PMID: 39083127 PMCID: PMC11291820 DOI: 10.1007/s12672-024-01203-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/29/2024] [Indexed: 08/03/2024] Open
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a common and fatal cancer, and its molecular mechanisms are still not fully understood. This study aimed to explore the potential molecular mechanisms and immune infiltration characteristics of celecoxib combined with sorafenib in the treatment of HCC by analyzing the differentially expressed genes (DEGs) from the GSE45340 dataset in the GEO database and identifying key genes. METHODS The GSE45340 dataset was downloaded from the GEO database, and DEGs were screened using GEO2R, and visualization and statistical analysis were performed. Metascape was used to perform functional annotation and protein-protein interaction network analysis of DEGs. The immune infiltration was analyzed using the TIMER database, and the expression of key genes and their relationship with patient survival were analyzed and verified using the UALCAN database. RESULTS A total of 2181 DEGs were screened through GEO2R analysis, and heat maps were drawn for the 50 genes with the highest expression. Metascape was used for enrichment analysis, and the enrichment results of KEGG and GO and the PPI network were obtained, and 44 core genes were screened. Analysis of the TIMER database found that 12 genes were closely related to tumor immune infiltration. UALCAN analysis further verified the differential expression of these genes in HCC and was closely related to the overall survival of patients. CONCLUSIONS Through comprehensive bioinformatics analysis, this study identified a group of key genes related to the treatment of HCC with celecoxib combined with sorafenib. These genes play an important role in tumor immune infiltration and patient survival, providing important clues for further studying the molecular mechanism of HCC and developing potential therapeutic targets.
Collapse
Affiliation(s)
- Wang Gu
- Hepatological Surgery Department, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei City, 230032, Anhui Province, China
| | - Dongyun Zeng
- Clinicopathological Diagnosis and Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China
| | - Chao Zhang
- Hepatological Surgery Department, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei City, 230032, Anhui Province, China.
| |
Collapse
|