1
|
Wang J, Chen Q, Shan Q, Liang T, Forde P, Zheng L. Clinical development of immuno-oncology therapeutics. Cancer Lett 2025; 617:217616. [PMID: 40054657 PMCID: PMC11930610 DOI: 10.1016/j.canlet.2025.217616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Immuno-oncology (IO) is one of the fastest growing therapeutic areas within oncology. IO agents work indirectly via the host's adaptive and innate immune system to recognize and eradicate tumor cells. Despite checkpoint inhibitors being only introduced to the market since 2011, they have become the second most approved product category. Current Food and Drug Administration (FDA)-approved classes of IO agents include: immune checkpoint inhibitors (ICIs), chimeric antigen receptor T-cell therapy (CAR-T), bi-specific T-cell engager (BiTE) antibody therapy, T-cell receptor (TCR) engineered T cell therapy, tumor-infiltrating lymphocyte (TIL) therapy, cytokine therapy, cancer vaccine therapy, and oncolytic virus therapy. Cancer immunotherapy has made progress in multiple cancer types including melanoma, non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), and urothelial carcinoma; however, several cancers remain refractory to immunotherapy. Future directions of IO include exploration in the neoadjuvant/perioperative setting, combination strategies, and optimizing patient selection through improved biomarkers.
Collapse
Affiliation(s)
- Jianxin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qi Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qiang Shan
- Department of General Surgery, Haining People's Hospital, Haining, 314400, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Patrick Forde
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
2
|
Ziogas DC, Theocharopoulos C, Aravantinou K, Boukouris AE, Stefanou D, Anastasopoulou A, Lialios PP, Lyrarakis G, Gogas H. Clinical benefit of immune checkpoint inhibitors in elderly cancer patients: Current evidence from immunosenescence pathophysiology to clinical trial results. Crit Rev Oncol Hematol 2025; 208:104635. [PMID: 39889861 DOI: 10.1016/j.critrevonc.2025.104635] [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: 06/27/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
The age-related decline in immunity appears to be associated not only with cancer development but also with differential responses to immune checkpoint inhibitors (ICIs). Despite their increasing utility across various malignancies and therapeutic settings, limited data -derived primarily from subgroup analyses of randomized controlled trials (RCTs), pooled meta-analyses, and retrospective studies- are available on the effects of aging on their efficacy and toxicity. Immunosenescence, characterized by the progressive decline of the function of the immune system, and inflammaging, a state of persistent low-grade sterile inflammation, may influence ICI outcomes. Additionally, the incidence, severity, and subtypes of immune-related adverse events (irAEs) may differ between older and younger individuals due to loss of immunotolerance. In the current review, starting from a a comprehensive discussion of the pathophysiology of immunosenescence, we proceed to critically review age-related retrospective and randomized evidence supporting FDA-approved ICIs. We highlight similarities or differences across age groups and the clinical benefit of ICIs in elderly versus younger cancer patients. The optimal integration of ICIs in geriatric oncology necessitates greater inclusion of this patient demographic in RCTs along with real-world data in order to acquire robust data which will guide evidence-based treatment decisions for this population.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Charalampos Theocharopoulos
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Katerina Aravantinou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Aristeidis E Boukouris
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Dimitra Stefanou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Amalia Anastasopoulou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Panagiotis-Petros Lialios
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - George Lyrarakis
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| |
Collapse
|
3
|
Chang Y, Long M, Shan H, Liu L, Zhong S, Luo JL. Combining gut microbiota modulation and immunotherapy: A promising approach for treating microsatellite stable colorectal cancer. Crit Rev Oncol Hematol 2025; 208:104629. [PMID: 39864533 DOI: 10.1016/j.critrevonc.2025.104629] [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: 12/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and lethal cancers worldwide, ranking third in incidence and second in mortality. While immunotherapy has shown promise in patients with deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H), its effectiveness in proficient mismatch repair (pMMR) or microsatellite stable (MSS) CRC remains limited. Recent advances highlight the gut microbiota as a potential modulator of anti-tumor immunity. The gut microbiome can significantly influence the efficacy of immune checkpoint inhibitors (ICIs), especially in pMMR/MSS CRC, by modulating immune responses and systemic inflammation. This review explores the role of the gut microbiota in pMMR/MSS CRC, the mechanisms by which it may enhance immunotherapy, and current strategies for microbiota modulation. We discuss the potential benefits of combining microbiota-targeting interventions with immunotherapy to improve treatment outcomes for pMMR/MSS CRC patients.
Collapse
Affiliation(s)
- Yujie Chang
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Min Long
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Hanguo Shan
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Logen Liu
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Shangwei Zhong
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Jun-Li Luo
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, USC, Hunan 410008, China.
| |
Collapse
|
4
|
Iwata Y, Tanaka C, Ohno S, Suetsugu T, Tanaka H, Watanabe T, Komori S, Nagao N, Katayama M, Kawai M. Real-world outcomes of stage II and III colorectal cancers treated by postoperative adjuvant chemotherapy based on the mismatch repair status. Surg Today 2025; 55:492-501. [PMID: 39249113 DOI: 10.1007/s00595-024-02932-9] [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: 06/13/2024] [Accepted: 07/31/2024] [Indexed: 09/10/2024]
Abstract
PURPOSE In Japan, immunohistochemistry for mismatch repair (MMR) proteins targeted at stage II and III colorectal cancers (CRCs) has been covered by national insurance since October, 2022. This study aimed to clarify the long-term outcomes of patients with stage II and III CRCs receiving postoperative adjuvant chemotherapy based on their MMR status. METHODS The outcomes of 640 patients who underwent radical surgery for stage II and III CRCs were analyzed retrospectively. RESULTS Deficient MMR (dMMR) was diagnosed in 41 (13.3%) patients with stage II and 28 (9.1%) patients with stage III CRC. The overall survival and recurrence rates were not significantly different between the patients with stage II and those with stage III CRC. The risk factors for recurrence among those with stage II CRC were tumors on the left side, T4 disease, and the presence of BRAF wild type. The recurrence rates were lower in the stage II CRC patients with sporadic dMMR than in those with suspected Lynch syndrome (LS). The first site of recurrence was more frequently the peritoneum or distant lymph node in patients with dMMR. CONCLUSIONS Stage II CRC patients with sporadic dMMR were found to have a very good prognosis. On the other hand, peritoneal dissemination or distant lymph node metastasis tended to develop in patients with dMMR.
Collapse
Affiliation(s)
- Yoshinori Iwata
- Department of Surgery, Gifu Prefectural General Medical Center, Gifu, Japan.
| | - Chihiro Tanaka
- Department of Surgery, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Shinya Ohno
- Department of Surgery, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Tomonari Suetsugu
- Department of Surgery, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Hideharu Tanaka
- Department of Surgery, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Taku Watanabe
- Department of Surgery, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Shuji Komori
- Department of Surgery, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Narutoshi Nagao
- Department of Surgery, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Masaki Katayama
- Department of Pathology, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Masahiko Kawai
- Department of Surgery, Gifu Prefectural General Medical Center, Gifu, Japan
| |
Collapse
|
5
|
Wu X, Mao Y, Xu N, Bai Y, Wang D, Chen X, Yin X, Deng Y, Yang J, Zhang J, Tang J, Huang Y, Li J, Luo S, Zheng H, Zhao W, Xu M, Li N, Mao Y, Gozman A, Xu J. Pembrolizumab in Patients of Chinese Descent with Microsatellite Instability-high/Mismatch Repair Deficient Advanced Solid Tumors: KEYNOTE-158 Final Analysis. Adv Ther 2025:10.1007/s12325-025-03142-6. [PMID: 40121391 DOI: 10.1007/s12325-025-03142-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/11/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION KEYNOTE-158 (NCT02628067) supported the US Food and Drug Administration approval of pembrolizumab for microsatellite instability-high (MSI-H)/mismatch repair deficient (dMMR) advanced solid tumors. Incidence of MSI-H/dMMR tumors in patients of Chinese descent is similar to that of Western populations. Cohort L of KEYNOTE-158 evaluated pembrolizumab in patients of Chinese descent with previously treated MSI-H/dMMR tumors. We previously reported an objective response rate (ORR) of 70% in 20 patients from cohort L which supported the approval in China of pembrolizumab in patients with MSI-H/dMMR solid tumors. Here we present results of the final analysis for 30 patients with median follow-up of 18 months. METHODS Eligible patients who had confirmed unresectable or metastatic MSI-H/dMMR tumors, and one or more prior lines of therapy, received 200 mg pembrolizumab Q3W (up to 35 cycles) until progression, toxicity, or withdrawal. Primary endpoint was ORR per RECIST 1.1 by central review. Secondary endpoints were duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety. RESULTS Thirty patients were enrolled; 7 (23%) were aged ≥ 65 years, and 20 (67%) were female. With median follow-up of 18 months, ORR was 66.7%. Median DOR was not reached (NR), with 12-month DOR rate of 85.9%. Median PFS was NR, with 18-month PFS rate of 63.0%. Median OS was NR, with 18-month OS rate of 78.8%. Treatment-related adverse events (AE) were reported in 22 (73%) patients. Grade 3-4 treatment-related AEs occurred in 7 (23%) patients. Immune-mediated AEs occurred in 11 (37%) patients of which 2 (7%) had grade 3 AEs. No grade ≥ 4 immune-mediated AEs occurred. CONCLUSION Pembrolizumab continues to provide clinically meaningful antitumor activity and durable responses with a manageable safety profile in patients of Chinese descent with MSI-H/dMMR advanced solid tumors. These results are consistent with those reported for patients in the global population and further support the use of pembrolizumab in patients of Chinese descent with MSI-H/dMMR tumors. TRIAL REGISTRATION NUMBER: NCT02628067.
Collapse
Affiliation(s)
- Xiaohua Wu
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yimin Mao
- Renji Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Nong Xu
- The First Affiliated Hospital Zhejiang University, Hangzhou, China
| | - Yuxian Bai
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Dong Wang
- Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaojun Chen
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | | | - Yanhong Deng
- The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jianwei Yang
- Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Jieqing Zhang
- Affiliated Cancer Hospital of Guangxi Medical University, Nanning, China
| | - Jie Tang
- Hunan Cancer Hospital, Changsha, China
| | - Yi Huang
- Hubei Cancer Hospital, Wuhan, China
| | - Jiayi Li
- The First Affiliated Hospital of Xiamen University-Oncology, Xiamen, China
| | - Suxia Luo
- Henan Cancer Hospital, Zhengzhou, China
| | | | - Weidong Zhao
- Anhui Provincial Hospital-Obstetrics and Gynecology, Hefei, China
| | | | - Nan Li
- MSD (China) Co., Ltd, Beijing, China
| | | | | | - Jianming Xu
- Fifth Medical Center of Chinese, PLA General Hospital, No. 8 Eastda Road, Fengtai District, Beijing, 100071, China.
| |
Collapse
|
6
|
Randrian V, Rousseau B. Combination of immune checkpoint blockade in metastatic colorectal cancers with microsatellite instability: A new standard of care? MED 2025; 6:100636. [PMID: 40088883 DOI: 10.1016/j.medj.2025.100636] [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: 02/15/2025] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 03/17/2025]
Abstract
Immunotherapy is highly efficient for the treatment of mismatch-repair-deficient metastatic colorectal cancer. Two recent articles1,2 challenge the current paradigm of anti-PD-1 monotherapy for the frontline treatment of metastatic mismatch-repair-deficient colorectal cancer, as the combination of an anti-PD-1 and anti-CTLA-4 showed improved response rate and progression-free survival compared to chemotherapy or anti PD-1.
Collapse
Affiliation(s)
- Violaine Randrian
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; CHU Poitiers, Université de Poitiers, Poitiers, France
| | | |
Collapse
|
7
|
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] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [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
|
8
|
Huang J, Xiao R, Shi S, Li Q, Li M, Xiao M, Wang Y, Yang Y, Li W, Tang Y. Circulating IL6 is involved in the infiltration of M2 macrophages and CD8+ T cells. Sci Rep 2025; 15:8681. [PMID: 40082587 PMCID: PMC11906812 DOI: 10.1038/s41598-025-92817-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
To elucidate the relationship between circulating cytokines and the prognosis of microsatellite-stable(MSS) colorectal cancer (CRC) patients, we examined the correlation between circulating cytokine levels and tumor immune infiltration microenvironment in this patient population. By conducting a preliminary analysis of the GEO database, we identified five core genes associated with colorectal cancer and further analyzed their impact on immune infiltration. We measured serum cytokine levels and validated the immune infiltration results through immunohistochemical staining of common inflammatory cell markers, including CD3, CD4, CD8, CD163, and FOXP3. Our findings indicate that serum cytokine levels significantly influence immune infiltration in colorectal cancer, particularly IL6 and IFNγ, which play crucial roles. Specifically, the infiltration of M2-type macrophages and CD8 + T cells is correlated with serum levels of IL6 and IFNγ. MSS CRC patients with elevated IL6 expression exhibit improved prognosis.
Collapse
Affiliation(s)
- Jing Huang
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Rui Xiao
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Suyujie Shi
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Qingshu Li
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Li
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Xiao
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yalan Wang
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaying Yang
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenwen Li
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China.
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China.
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Yi Tang
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China.
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China.
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
9
|
Sanghvi G, R R, Kashyap A, Sabarivani A, Ray S, Bhakuni PN. Identifying the function of kinesin superfamily proteins in gastric cancer: Implications for signal transduction, clinical significance, and potential therapeutic approaches. Clin Res Hepatol Gastroenterol 2025; 49:102571. [PMID: 40064398 DOI: 10.1016/j.clinre.2025.102571] [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: 01/18/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
Gastric cancer (GC), a leading cause of cancer-related mortality, poses a significant global health challenge. Given its complex etiology, understanding the molecular pathways driving GC progression is crucial for developing innovative therapeutic strategies. Among the diverse proteins involved in cellular transport and mitotic regulation, kinesin superfamily proteins (KIFs) have emerged as key players in tumor biology. These motor proteins mediate intracellular transport along microtubules and are essential for processes such as cell division, signaling, and organelle distribution. Evidence indicates that specific KIFs are dysregulated in GC, potentially driving cancer cell proliferation, metastasis, and chemoresistance. Moreover, aberrant KIF expression has been associated with poorer prognoses, highlighting their potential as biomarkers for early diagnosis and therapeutic intervention. This review explores the roles of KIFs in GC and assesses their implications for research and clinical applications. By elucidating the significance of KIFs in GC, this discussion aims to inspire novel insights in cancer biology and advance targeted therapeutic strategies.
Collapse
Affiliation(s)
- Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India
| | - A Sabarivani
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003, India
| | - Pushpa Negi Bhakuni
- Department of Allied Science, Graphic Era Hill University, Bhimtal, Uttarakhand 248002, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India.
| |
Collapse
|
10
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
11
|
Fischer AK, Kroesen A, Büttner R. [Secondary malignant neoplasms with underlying Lynch syndrome and coincidental ulcerative colitis]. CHIRURGIE (HEIDELBERG, GERMANY) 2025:10.1007/s00104-025-02251-w. [PMID: 40029372 DOI: 10.1007/s00104-025-02251-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 03/05/2025]
Affiliation(s)
- Anne Kristin Fischer
- Institut für Pathologie, Universität zu Köln, Kerpener Str. 62, 50937, Köln, Deutschland.
| | - Anton Kroesen
- Krankenhaus Porz am Rhein, Urbacher Weg 19, 51149, Köln, Deutschland
| | - Reinhard Büttner
- Institut für Pathologie, Universität zu Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| |
Collapse
|
12
|
Bari S, Matejcic M, Kim RD, Xie H, Sahin IH, Powers BD, Teer JK, Chan TA, Felder SI, Schmit SL. Practice Patterns and Survival Outcomes of Immunotherapy for Metastatic Colorectal Cancer. JAMA Netw Open 2025; 8:e251186. [PMID: 40111368 PMCID: PMC11926646 DOI: 10.1001/jamanetworkopen.2025.1186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/14/2025] [Indexed: 03/22/2025] Open
Abstract
Importance Immune checkpoint inhibitors (ICIs) have been approved for treatment of microsatellite instable (MSI-H) metastatic colorectal cancer (mCRC), but factors associated with receipt and efficacy of ICIs in routine clinical practice remain largely unknown. Objective To identify factors associated with receipt of ICIs and associated survival outcomes among patients with mCRC in routine clinical practice. Design, Setting, and Participants This population-based cohort study used deidentified data from a nationwide electronic health record-derived database to include 18 932 patients diagnosed with mCRC between January 2013 and June 2019. Population-based patients were diagnosed with de novo mCRC and had at least 2 documented clinical visits on or after the date of diagnosis. The study analyses were performed between September 2020 and April 2021. Exposure Patients received ICI therapy and/or chemotherapy as part of a systemic treatment for mCRC. Main Outcomes and Measures The outcomes were receipt of ICI therapy, overall survival (OS), and time to treatment discontinuation (TTD). Results In this cohort study of 18 932 patients diagnosed with mCRC (10 537 [55.7%] male; 546 [2.9%] Asian, 2005 [10.6%] Black or African American, 1674 [8.8%] Hispanic, 12 338 [65.2%] White, 4043 [21.4%] unknown race or ethnicity; median [IQR] age at metastatic diagnosis, 64.6 [55.0-73.3] years), patients with MSI-H tumors had a significantly higher probability of receiving ICIs than those with microsatellite stable (MSS) tumors (odds ratio [OR], 22.66 [95% CI, 17.30-29.73]; P < .001), whereas patients initially diagnosed with synchronous mCRC had significantly lower odds of receiving ICIs than patients with metachronous mCRC (OR, 0.57 [95% CI, 0.45-0.73]; P < .001). Patients with MSI-H tumors who received ICIs as first line of therapy had significantly longer OS than those receiving chemotherapy only (HR, 0.37 [95% CI, 0.25-0.56]; P < .001). Among patients with MSS tumors, ICI-based therapy was associated with significantly longer OS for patients with high albumin level (vs low: HR, 0.28 [95% CI, 0.18-0.45]; P < .001) and antibiotic use (vs nonuse: HR, 0.43 [95% CI, 0.27-0.67]; P < .001), but a significantly shorter OS for patients with synchronous mCRC (vs metachronous: HR, 1.90 [95% CI, 1.24-2.89]; P = .003). In addition, 29 out of 235 patients with MSS tumors (12.3%) experienced durable responses on ICI-based therapy. Similar patterns of associations with TTD were observed. Conclusions and Relevance In this cohort study of patients with mCRC, clinical characteristics were associated with different survival outcomes in patients treated with ICI-based therapy, with important clinical implications for patients with MSS tumors who are generally unresponsive to immunotherapy.
Collapse
Affiliation(s)
- Shahla Bari
- Department of Medical Oncology, Duke Cancer Institute, Durham, North Carolina
| | - Marco Matejcic
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Richard D Kim
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hao Xie
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Benjamin D Powers
- Department of Surgery, University of Maryland School of Medicine, Baltimore
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio
| | - Seth I Felder
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Stephanie L Schmit
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
13
|
Kaczorowski M, Chłopek M, Daum O, Ylaya K, Vaněček T, Szczepaniak M, Krawczyk K, Kowalik A, Michal M, Lasota J, Miettinen M. MMR deficiency is frequent in colorectal carcinomas with diffuse SLFN11 immunostaining: clinicopathologic and molecular study of 31 cases identified among 3,300 tumors. J Pathol Clin Res 2025; 11:e70025. [PMID: 40105034 PMCID: PMC11920882 DOI: 10.1002/2056-4538.70025] [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: 12/23/2024] [Revised: 02/04/2025] [Accepted: 02/25/2025] [Indexed: 03/20/2025]
Abstract
Schlafen 11 (SLFN11), a regulator of cell fate following DNA injury, sensitizes tumor cells to DNA-damaging agents. Patients with SLFN11-positive tumors may benefit from DNA-damaging chemotherapies. SLFN11 has been studied in different types of cancer including colorectal carcinomas. However, colorectal carcinomas with diffuse positivity (expression in ≥80% of tumor cells) have not been meticulously characterized. SLFN11 immunostaining of tumor microarrays (TMAs) with 3,300 primary CRCs identified 65 (~2.0%) tumors with focal staining (<10% of tumor nuclei positive), 83 (~2.5%) with patchy (≥10% and <80%) and 51 (~1.5%) with diffuse (≥80%) SLFN11 positivity. The latter was confirmed on full sections from donor blocks in 31 (~1%) cases, which were further studied including evaluation of additional immunohistochemical markers, genotyping with targeted DNA sequencing, and assessment of microsatellite instability. SLFN11-positive carcinomas were mostly (21/31, 68%) right-sided tumors with a female predominance (21/31, 68%) and median age of 67 years. Eighteen of 31 (58%) contained areas of mucinous differentiation. Deficiency of mismatch repair proteins was detected in 65% (20/31) of SLFN11-positive carcinomas. Moreover, MLH1 (n = 2), MSH2, MSH6, and PMS2 germline mutations were identified in 25% (5/20) of patients with mismatch repair deficient tumors. BRAF p.V600E mutation was found in 45% (9/20) of mismatch repair deficient, but only 1 of 11 proficient tumors. Colorectal carcinomas with diffuse SLFN11 positivity were often mismatch repair deficient tumors with their typical clinical, morphological, and molecular characteristics.
Collapse
Affiliation(s)
- Maciej Kaczorowski
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
- Department of Clinical and Experimental Pathology, Wrocław Medical University, Wrocław, Poland
| | | | - Ondřej Daum
- Sikl's Institute of Pathology, Faculty of Medicine and Teaching Hospital in Plzen, Charles University, Plzen, Czech Republic
- Bioptical Laboratory, Ltd., Plzen, Czech Republic
| | - Kris Ylaya
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Tomáš Vaněček
- Sikl's Institute of Pathology, Faculty of Medicine and Teaching Hospital in Plzen, Charles University, Plzen, Czech Republic
- Bioptical Laboratory, Ltd., Plzen, Czech Republic
| | | | - Karol Krawczyk
- Department of Molecular Diagnostics, Holycross Cancer Center, Kielce, Poland
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holycross Cancer Center, Kielce, Poland
| | - Michal Michal
- Sikl's Institute of Pathology, Faculty of Medicine and Teaching Hospital in Plzen, Charles University, Plzen, Czech Republic
- Bioptical Laboratory, Ltd., Plzen, Czech Republic
| | - Jerzy Lasota
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Markku Miettinen
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| |
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
|
Argilés G, Arnold D, Cervantes A. Anti-PD-1 treatment for MSI-H/MMRD tumors. A journey from genomics to transformative patient breakthroughs. Ann Oncol 2025; 36:231-232. [PMID: 39984220 DOI: 10.1016/j.annonc.2025.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/23/2025] Open
Affiliation(s)
- G Argilés
- Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, U.S..
| | - D Arnold
- Asklepios Tumorzentrum Hamburg, AK Altona, Hamburg, Germany
| | - A Cervantes
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, University of Valencia, Spain
| |
Collapse
|
16
|
Jiao J, Wu Y, Wu S, Jiang J. Enhancing Colorectal Cancer Treatment Through VEGF/VEGFR Inhibitors and Immunotherapy. Curr Treat Options Oncol 2025; 26:213-225. [PMID: 40045029 DOI: 10.1007/s11864-025-01306-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2025] [Indexed: 03/20/2025]
Abstract
OPINION STATEMENT Colorectal cancer, ranking as the third most prevalent malignancy globally, substantially benefits from both immunotherapy and VEGF/VEGFR inhibitors. Nevertheless, the use of monotherapy proves inadequate in effectively tackling the heterogeneity of tumors and the intricacies of their microenvironment, frequently leading to drug resistance and immune evasion. This situation underscores the pressing need for innovative strategies aimed at augmenting the effectiveness and durability of treatments. Clinical research demonstrates that the combination of VEGF/VEGFR inhibitors (primarily including VEGF/VEGFR-targeted drugs and multi-kinase inhibitors) with immune checkpoint inhibitors creates a synergistic effect in the treatment of colorectal cancer. Our analysis explores how VEGF/VEGFR inhibitors recalibrate the tumor microenvironment, modulate immune cell functions, and influence the expression of immune checkpoints and cytokines. Furthermore, we critically evaluate the preclinical and clinical feasibility of these combined therapeutic approaches. Despite the potential for toxicity, the significant benefits and prospective applications of these strategies warrant thorough exploration. Exploring the synergistic mechanisms of these combined treatments has the potential to inaugurate a new paradigm in oncology, enabling more personalized and efficacious treatment modalities. Additionally, the synergy between VEGF/VEGFR inhibitors and nascent immunotherapies emerges as a promising field of inquiry.
Collapse
Affiliation(s)
- Jing Jiao
- Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Tumor Biological Treatment, Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University Jiangsu Engineering Research Center for Tumor Immunotherapy, Soochow University, Juqian Road №185, Changzhou, 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - You Wu
- Department of Tumor Biological Treatment, Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University Jiangsu Engineering Research Center for Tumor Immunotherapy, Soochow University, Juqian Road №185, Changzhou, 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University Jiangsu Engineering Research Center for Tumor Immunotherapy, Soochow University, Juqian Road №185, Changzhou, 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Jingting Jiang
- Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Department of Tumor Biological Treatment, Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University Jiangsu Engineering Research Center for Tumor Immunotherapy, Soochow University, Juqian Road №185, Changzhou, 213003, China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|
17
|
Calegari MA, Zurlo IV, Dell'Aquila E, Basso M, Orlandi A, Bensi M, Camarda F, Anghelone A, Pozzo C, Sperduti I, Salvatore L, Santini D, Corsi DC, Bria E, Tortora G. Chemotherapy Rechallenge or Reintroduction Compared to Regorafenib or Trifluridine/Tipiracil for Pretreated Metastatic Colorectal Cancer Patients: A Propensity Score Analysis of Treatment Beyond Second Line (Proserpyna Study). Clin Colorectal Cancer 2025; 24:1-10.e4. [PMID: 38969549 DOI: 10.1016/j.clcc.2024.06.002] [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: 03/12/2023] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND The optimal treatment for metastatic colorectal cancer (mCRC) beyond second line is still questioned. Besides the standard of care agents (regorafenib, REG, or trifluridine/tipiracil, FTD/TPI), chemotherapy rechallenge or reintroduction (CTr/r) are commonly considered in clinical practice, despite weak supporting evidence. The prognostic performance of CTr/r, REG and FTD/TPI in this setting are herein evaluated. PATIENTS AND METHODS PROSERpYNa is a multicenter, observational, retrospective study, in which patients with refractory mCRC, progressing after at least 2 lines of CT, treated with CTr/r, REG or FTD/TPI, are considered eligible and were enrolled in 2 independent data sets (exploratory and validation). Primary endpoint was overall survival (OS); secondary endpoints were investigator-assessed progression-free survival (PFS), objective response rate (RR) and safety. A propensity score adjustment was accomplished for survival analyses. RESULTS Data referring to patients treated between Jan-10 and Jan-19 from 3 Italian institutions were gathered (341 and 181 treatments for exploratory and validation data sets respectively). In the exploratory cohort, median OS (18.5 vs. 6.5 months), PFS (6.1 vs. 3.5 months) and RR (28.6% vs. 1.4%) were significantly longer for CTr/r compared to REG/FTD/TPI. Survival benefits were retained at the propensity score analysis, adjusted for independent prognostic factors identified at multivariate analysis. Moreover, these results were confirmed within the validation cohort analyses. CONCLUSIONS Although the retrospective fashion, CTr/r proved to be a valuable option in this setting in a real-world context, providing superior outcomes compared to standard of care agents at the price of a moderate toxicity.
Collapse
Affiliation(s)
- M A Calegari
- Comprehensive Cancer Center, UOC Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.
| | - I V Zurlo
- Medical Oncology, Ospedale San Giovanni Calibita Fatebenefratelli, Roma, Italy
| | - E Dell'Aquila
- Department of Medical Oncology, Campus Biomedico University, Rome, Italy; Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - M Basso
- Comprehensive Cancer Center, UOC Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - A Orlandi
- Comprehensive Cancer Center, UOC Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - M Bensi
- Comprehensive Cancer Center, UOC Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy; Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - F Camarda
- Comprehensive Cancer Center, UOC Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy; Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - A Anghelone
- Comprehensive Cancer Center, UOC Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy; Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - C Pozzo
- Comprehensive Cancer Center, UOC Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - I Sperduti
- Biostatistics, IRCCS Regina Elena National Cancer Institute, Roma, Italy
| | - L Salvatore
- Comprehensive Cancer Center, UOC Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy; Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - D Santini
- Department of Medical Oncology, Campus Biomedico University, Rome, Italy; Department of Radiology, Oncology and Pathology, Policlinico Umberto, I Sapienza University of Rome, Rome, Italy
| | - D C Corsi
- Medical Oncology, Ospedale San Giovanni Calibita Fatebenefratelli, Roma, Italy
| | - E Bria
- Comprehensive Cancer Center, UOC Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy; Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - G Tortora
- Comprehensive Cancer Center, UOC Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy; Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
18
|
Nie H, Fang S, Zhou R, Jia Y, Zhou J, Ning Y, Yu Y, Hong Y, Xu F, Zhao Q, Nie J, Wang F. Upregulation of RIG-I is Critical for Responsiveness to IFN-α Plus Anti-PD-1 in Colorectal Cancer. Cancer Med 2025; 14:e70802. [PMID: 40116486 PMCID: PMC11926914 DOI: 10.1002/cam4.70802] [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: 02/03/2025] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUNDS Immunotherapy is a promising and effective approach that has achieved significant curative effects in colorectal cancer (CRC). Recently, retinoic acid-inducible gene I (RIG-I) has been shown to play a critical role in tumor immunity. However, the correlation between RIG-I and immunotherapy in CRC remains unclear. METHODS RIG-I expression was measured in CRC and normal samples based on analysis of the public databases, a tissue microarray, and CRC cell lines. The correlation between RIG-I and immune microenvironment was explored using well-established biological algorithms and in vitro and in vivo experiments. RESULTS We discovered that RIG-I expression was downregulated in CRC compared with normal samples. The bioinformatic algorithms indicated that high RIG-I-expressing samples showed a positive correlation with IFN-α response and enrichment of antitumor immune cells, especially CD8+ T cells. Furthermore, knockdown of RIG-I expression efficiently reduced the cell death, STAT1 phosphorylation, and CXCL10/11 expression induced by IFN-α in CRC cells. Finally, an in vivo study showed that the infiltration of CD3+ CD8+ T cells was significantly decreased in the RIG-I knockout group. An animal model further confirmed that the inhibition of tumor growth induced by IFN-α plus anti-PD-1 therapy was dependent on RIG-I expression. CONCLUSION RIG-I is a promising biomarker for CRC immunotherapy, which provides a novel concept for combinatorial immunotherapy.
Collapse
Affiliation(s)
- Haihang Nie
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Shilin Fang
- Department of Infectious DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Rui Zhou
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yifan Jia
- Department of PainRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jingkai Zhou
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yumei Ning
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yali Yu
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yuntian Hong
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Fei Xu
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Qiu Zhao
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jiayan Nie
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Fan Wang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
19
|
Gordon AC, Zahra SA, Serhal M, Kircher SM, Kalyan A, Sato K, Riaz A, Hohlastos E, Salem R, Lewandowski RJ. Escalated Segmental and Modified Radiation Lobectomy Dosing for Yttrium-90 Radioembolization of Liver-Dominant Metastatic Colorectal Cancer: 10-year Outcomes. Clin Colorectal Cancer 2025:S1533-0028(25)00026-X. [PMID: 40133187 DOI: 10.1016/j.clcc.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE This study evaluates the safety and efficacy of escalated-dosing Yttrium-90 transarterial radioembolization (TARE) for unresectable, unablatable metastatic colorectal cancer (mCRC) to the liver. MATERIALS AND METHODS A retrospective review (September 2009 to March 2020) included 45 patients with liver-dominant mCRC treated with segmental Y90 or modified radiation lobectomy. Patient demographics, treatment details, adverse events, imaging response, and overall survival (OS) were analyzed. OS Prognosticators were examined using log-rank test and Cox proportional hazards regression. RESULTS 45 patients (median age 61.4 years; 60% male) were included, with 96% ECOG 0-1. Prior treatments included primary site resection (93%), liver resection (65%), chemotherapy (60%), and ablation (27%). Extrahepatic disease was present in 51%. 71% of patients had < 25% liver tumor burden (mean tumor size = 4.8 cm). Treatment was technically successful in all cases, with 4% 30-day mortality. Adverse events were mostly low-grade, including fatigue (58%) and abdominal pain (20%). Mean neutrophil-to-lymphocyte ratio (NLR) increase was 2.9, and 33% of patients showed 50% reduction in CEA. Imaging responses (RECIST) included SD (80%), PR (18%), PD (2%), and CR (0%), with PET/CT showing 39% objective response after 4.2 months. Median OS was 41.9 months (95% CI 15.4-NE). Extrahepatic disease significantly reduced OS (15.7 vs. 44.4 months, P = .0033). Both pre- and post-NLR (HR:1.42, P = .007; HR 1.12, P = .027) were associated with worse OS. In the multivariable analysis, Pre-NLR and extrahepatic disease remained adverse prognosticators. CONCLUSION Y90 TARE with escalated dosing demonstrated an acceptable safety profile in heavily pretreated mCRC patients. Extrahepatic disease and pre-NLR were significant adverse prognosticators. Future studies should explore Y90 TARE dosing in mCRC patients.
Collapse
Affiliation(s)
- Andrew C Gordon
- Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, IL
| | - Saad Abu Zahra
- Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, IL
| | - Muhamad Serhal
- Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, IL
| | - Sheetal M Kircher
- Department of Medicine, Division of Hematology and Oncology, Northwestern University, Chicago, IL
| | - Aparna Kalyan
- Department of Medicine, Division of Hematology and Oncology, Northwestern University, Chicago, IL
| | - Kent Sato
- Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, IL
| | - Ahsun Riaz
- Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, IL
| | - Elias Hohlastos
- Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, IL
| | - Riad Salem
- Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, IL; Department of Medicine, Division of Gastroenterology and Hepatology, Northwestern University, Chicago, IL; Department of Surgery, Division of Transplant Surgery, Northwestern University, Chicago, IL
| | - Robert J Lewandowski
- Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, IL; Department of Medicine, Division of Gastroenterology and Hepatology, Northwestern University, Chicago, IL; Department of Surgery, Division of Transplant Surgery, Northwestern University, Chicago, IL.
| |
Collapse
|
20
|
Hecht JR, Michot JM, Bajor D, Patnaik A, Chung KY, Wang J, Falchook G, Cleary JM, Kim R, Krishnamurthy A, Marathe O, Youssoufian H, Ellis C, Waszak A, Ghosh S, Zhang H, Yablonski K, Shah SD, Diaz-Padilla I, Ulahannan S. CITRINO: phase 1 dose escalation study of anti-LAG-3 antibody encelimab alone or in combination with anti-PD-1 dostarlimab in patients with advanced/metastatic solid tumours. BJC REPORTS 2025; 3:10. [PMID: 40016550 PMCID: PMC11868598 DOI: 10.1038/s44276-024-00118-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 03/01/2025]
Abstract
BACKGROUND Dual programmed cell death protein (ligand)-1 (PD-[L]1) and lymphocyte-activation gene-3 (LAG-3) blockade has demonstrated improved anti-tumour response in some advanced solid tumours. CITRINO, a two-part, Phase 1 dose-escalation study, evaluated encelimab (TSR-033; novel anti-LAG-3) monotherapy and in combination in patients with advanced/metastatic solid tumours. METHODS Part 1 (P1) involved dose escalation (20-720 mg Q2W) of encelimab as monotherapy (P1A/B) and with dostarlimab (500 mg Q3W) in patients with previously treated advanced/metastatic solid tumours (P1C). P2 involved cohort expansion in patients with anti-PD-(L)1-naïve microsatellite stable advanced/metastatic colorectal cancer with recommended phase 2 dose (RP2D) of encelimab with dostarlimab as third/fourth-line therapy (P2A), or with dostarlimab, bevacizumab and mFOLFOX6/FOLFIRI as second-line therapy (P2B). Objectives included RP2D, safety/tolerability, efficacy, pharmacokinetics/pharmacodynamics, and exploratory biomarkers. RESULTS Maximum tolerated encelimab dose was not reached; 720 mg Q2W was used for P2 plus dostarlimab 1000 mg Q6W. One dose-limiting toxicity occurred (Grade 2 myasthenia gravis; P1A). No clinical responses were observed in P1; 1 (3%) and 4 (17%) patients achieved partial response in P2A and 2B, respectively. CONCLUSIONS Encelimab has a manageable safety profile as a monotherapy and in tested combinations; however, anti-tumour activity was limited. CLINICAL TRIAL REGISTRATION NCT03250832.
Collapse
Affiliation(s)
- J Randolph Hecht
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
| | - Jean-Marie Michot
- Département d'Innovation Thérapeutique et d'Essais Précoces, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - David Bajor
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | | | - Ki Y Chung
- Department of Medicine, Prisma Health Cancer Institute, Greenville, SC, USA
| | - Judy Wang
- Drug Development Unit, Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, FL, USA
| | - Gerald Falchook
- Drug Development Unit, Sarah Cannon Research Institute at HealthONE, Denver, CO, USA
| | - James M Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Richard Kim
- Department of Medical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | - Omkar Marathe
- The Oncology Institute of Hope and Innovation, Whittier, CA, USA
| | | | | | - Angela Waszak
- Oncology Clinical Development, GSK, Waltham, MA, USA
| | | | - Hailei Zhang
- Oncology Clinical Development, GSK, Waltham, MA, USA
| | | | - Shruti D Shah
- Oncology Clinical Development, GSK, Waltham, MA, USA
| | | | - Susanna Ulahannan
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Sarah Cannon Research Institute, Nashville, TN, USA
| |
Collapse
|
21
|
Tang WW, Battistone B, Bauer KM, Weis AM, Barba C, Fadlullah MZH, Ghazaryan A, Tran VB, Lee SH, Agir ZB, Nelson MC, Victor ES, Thibeaux A, Hernandez C, Tantalla J, Tan AC, Rao D, Williams M, Drummond MJ, Beswick EJ, Round JL, Ekiz HA, Voth WP, O'Connell RM. A microRNA-regulated transcriptional state defines intratumoral CD8 + T cells that respond to immunotherapy. Cell Rep 2025; 44:115301. [PMID: 39951377 PMCID: PMC11924119 DOI: 10.1016/j.celrep.2025.115301] [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: 08/04/2024] [Revised: 11/24/2024] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
The rising incidence of advanced-stage colorectal cancer (CRC) and poor survival outcomes necessitate new and effective therapies. Immune checkpoint inhibitors (ICIs), specifically anti-PD-1 therapy, show promise, yet clinical determinants of a positive response are suboptimal. Here, we identify microRNA-155 (miR-155) as necessary for CD8+ T cell-infiltrated tumors through an unbiased in vivo CRISPR-Cas9 screen identifying functional tumor antigen-specific CD8+ T cell-expressed microRNAs. T cell miR-155 is required for anti-PD-1 responses and for a vital intratumor CD8+ T cell differentiation cascade by repressing Ship-1, inhibiting Tcf-1 and stemness, and subsequently enhancing Cxcr6 expression, anti-tumor immunity, and effector functions. Based on an underlying miR-155-dependent CD8+ T cell transcriptional profile, we identify a gene signature that predicts ICI responses across 12 diverse cancers. Together, our findings support a model whereby miR-155 serves as a central regulator of CD8+ T cell-dependent cancer immunity and ICI responses that may be leveraged for future therapeutics.
Collapse
Affiliation(s)
- William W Tang
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Ben Battistone
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Kaylyn M Bauer
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Allison M Weis
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Cindy Barba
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Muhammad Zaki Hidayatullah Fadlullah
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Arevik Ghazaryan
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Van B Tran
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Soh-Hyun Lee
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Z Busra Agir
- Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey
| | - Morgan C Nelson
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Emmanuel Stephen Victor
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Amber Thibeaux
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Colton Hernandez
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Jacob Tantalla
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Aik C Tan
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Dinesh Rao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matthew Williams
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Micah J Drummond
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT 84108, USA
| | - Ellen J Beswick
- Division of Digestive Disease and Nutrition, Department of Internal Medicine, University of Kentucky, Lexington, KY 40508, USA
| | - June L Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - H Atakan Ekiz
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Warren P Voth
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
22
|
Zhang Y, Li H, Zhao Y, Liu L, Zhou Y, Pan X, Ding Y, Liao W, Qi L, Huang C, Tang N. Macrocarpal I induces immunogenic cell death and synergizes with immune checkpoint inhibition by targeting tubulin and PARP1 in colorectal cancer. Cell Death Discov 2025; 11:73. [PMID: 39987121 PMCID: PMC11846858 DOI: 10.1038/s41420-025-02360-9] [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: 06/19/2024] [Revised: 01/26/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025] Open
Abstract
Colorectal cancer (CRC) presents an obstacle to immunotherapy, primarily because most cases are microsatellite stable (MSS) tumors, which are often described as "cold tumors" with limited immunogenicity. Recent studies have indicated that several therapeutic approaches, such as chemotherapy and targeted therapies, can elicit immunogenic cell death (ICD) and stimulate immune responses. However, challenges such as target affinity and in vivo pharmacokinetics limit the efficacy and immune response of current targeted therapies. In this study, we demonstrate that Macrocarpal I is a potent inducer of ICD by activating the PERK/eIF2A/ATF4/CHOP signaling pathway. Furthermore, Macrocarpal I induces apoptosis and ferroptosis, both of which act as triggers for ICD. Mechanistically, Macrocarpal I directly targets TUBB2B and PARP1, disrupting microtubule polymerization and DNA repair processes. Importantly, treatment with Macrocarpal I enhances the anti-tumor immune response and augments responsiveness to anti-PD-1 therapy in an MC38 syngeneic mouse model of CRC.
Collapse
Affiliation(s)
- Yaxin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Huali Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yali Zhao
- Department of Pathology, Shenzhen Longgang Central Hospital, Shenzhen, 518100, China
| | - Lingtao Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yi Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xingyan Pan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yanqing Ding
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wenting Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Lu Qi
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Chengmei Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Na Tang
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, 518020, China.
| |
Collapse
|
23
|
Li Y, Liu F, Cai Q, Deng L, Ouyang Q, Zhang XHF, Zheng J. Invasion and metastasis in cancer: molecular insights and therapeutic targets. Signal Transduct Target Ther 2025; 10:57. [PMID: 39979279 PMCID: PMC11842613 DOI: 10.1038/s41392-025-02148-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
The progression of malignant tumors leads to the development of secondary tumors in various organs, including bones, the brain, liver, and lungs. This metastatic process severely impacts the prognosis of patients, significantly affecting their quality of life and survival rates. Research efforts have consistently focused on the intricate mechanisms underlying this process and the corresponding clinical management strategies. Consequently, a comprehensive understanding of the biological foundations of tumor metastasis, identification of pivotal signaling pathways, and systematic evaluation of existing and emerging therapeutic strategies are paramount to enhancing the overall diagnostic and treatment capabilities for metastatic tumors. However, current research is primarily focused on metastasis within specific cancer types, leaving significant gaps in our understanding of the complex metastatic cascade, organ-specific tropism mechanisms, and the development of targeted treatments. In this study, we examine the sequential processes of tumor metastasis, elucidate the underlying mechanisms driving organ-tropic metastasis, and systematically analyze therapeutic strategies for metastatic tumors, including those tailored to specific organ involvement. Subsequently, we synthesize the most recent advances in emerging therapeutic technologies for tumor metastasis and analyze the challenges and opportunities encountered in clinical research pertaining to bone metastasis. Our objective is to offer insights that can inform future research and clinical practice in this crucial field.
Collapse
Affiliation(s)
- Yongxing Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Graduate School of Biomedical Science, Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lijun Deng
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
24
|
Ying L, Zhang L, Chen Y, Huang C, Zhou J, Xie J, Liu L. Predicting immunotherapy prognosis and targeted therapy sensitivity of colon cancer based on a CAF-related molecular signature. Sci Rep 2025; 15:6387. [PMID: 39984646 PMCID: PMC11845748 DOI: 10.1038/s41598-025-90899-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/17/2025] [Indexed: 02/23/2025] Open
Abstract
The role of cancer-associated fibroblasts (CAFs) in modulating the tumor microenvironment (TME) is gaining attention, yet their impact on prognosis and therapeutic response in colon cancer remains unclear. Here, we identified genes associated with CAF infiltration via weighted gene co-expression network analysis (WGCNA) utilizing data from The Cancer Genome Atlas (TCGA) and GSE39582 cohorts. Univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression analyses were used to construct CAF molecular signatures (CAFscore). Patients were categorized into high and low CAFscore groups to analyze clinicopathological traits, somatic mutations, immune evasion, and treatment responses. In this study, a total of 244 genes were correlated with CAF infiltration, with 11 linked to overall survival. Notably, FSTL3, CRIP2, and SLC2A3 were selected for the CAFscore. A higher CAFscore was associated with poorer prognoses, increased malignancy, and therapeutic resistance, particularly among patients with high tumor mutation burden and microsatellite instability. Furthermore, elevated FSTL3 expression was associated with reduced CD8+ T cell infiltration, indicating a suppressive TME. Mechanistically, CAFs may promote immune evasion via NAMPT ligand-receptor interactions based on single-cell RNA sequencing data. Thus, the CAFscore is crucial for personalizing treatment strategies and identifying patients who require more aggressive management.
Collapse
Affiliation(s)
- Leqian Ying
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Lu Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Yanping Chen
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Chunchun Huang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Jingyi Zhou
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Jinbing Xie
- Department of Radiology, Nurturing Center of Jiangsu Province for the State Laboratory of AI Imaging and Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Lin Liu
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China.
| |
Collapse
|
25
|
Lee SH, Pankaj A, Yilmaz O, Deshpande V, Yilmaz O. Beta-2-microglobulin positive tumor cells and CD8 positive lymphocytes are associated with outcome in post-neoadjuvant colorectal cancer resections. Hum Pathol 2025; 155:105737. [PMID: 39988058 DOI: 10.1016/j.humpath.2025.105737] [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: 12/30/2024] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Locally advanced colorectal cancers are treated with neoadjuvant therapy (NAT), which has been shown to alter the characteristics of the tumor including size, lymph node yield, and histologic grade. We seek to interrogate the effect of NAT on the immune microenvironment. We compared 190 patients with colorectal adenocarcinoma treated with NAT with those without NAT (n = 926). We evaluated clinicopathologic and molecular factors and performed immunohistochemistry and quantification on tissue microarrays for HLA class I/II proteins, beta-2-microglobulin (B2M), CD8, CD163, LAG3, PD-L1, and FoxP3. Patients in the NAT group were younger (60.9 vs 67.9, p < 0.001) and more often male (59.5 vs. 47.9, p = 0.004) than those in the non-NAT group. Tumors in the NAT group were smaller (3.5 vs 4.7 cm, p < 0.001), less often high grade (6.5% vs. 16.2%, p = 0.001), more frequently in the rectum (68.9% vs. 6.6%, p < 0.001) and associated with lower lymph node yields (p = 0.002); however, the incidence of extramural venous invasion, perineural invasion, and AJCC stage 3-4 disease were not different. Immune cells positive for CD8 (p = 0.011) were significantly lower in the NAT group. A high number of CD8+ cells and higher expression of B2M in tumor cells showed a significant survival benefit in both NAT and non-NAT group. NAT is associated with an immune-low tumor environment. CD8+ cells and tumor B2M expression may help identify a subset of immune high-tumors following NAT. This identification could aid in determining patients who may benefit from conservative management of colorectal carcinomas.
Collapse
Affiliation(s)
- Soo Hyun Lee
- Department of Pathology, Boston Medical Center, Boston, MA, USA
| | - Amaya Pankaj
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Omer Yilmaz
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Vikram Deshpande
- Harvard Medical School, Boston, MA, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Osman Yilmaz
- Harvard Medical School, Boston, MA, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
26
|
Li J, Zhou X, Wu L, Ma J, Tan Y, Wu S, Zhu J, Wang Q, Shi Q. Optimal early endpoint for second-line or subsequent immune checkpoint inhibitors in previously treated advanced solid cancers: a systematic review. BMC Cancer 2025; 25:293. [PMID: 39966752 PMCID: PMC11837729 DOI: 10.1186/s12885-025-13712-0] [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: 06/01/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND The administration of second-line or subsequent immune checkpoint inhibitors (ICIs) in previously treated patients with advanced solid cancers has been clinically investigated. However, previous clinical trials lacked an appropriate primary endpoint for efficacy assessment. This systematic review aimed to explore the most optimal early efficacy endpoint for such trials. METHODS Phase 2 or 3 clinical trials involving patients with advanced solid cancers with disease progression following standard first-line therapy receiving second-line or subsequent ICI administration, with adequate survival outcome data, were included from PubMed, Embase, Web of Science, and Cochrane Library databases before February 2023. Quality assessment was conducted using the Cochrane tool and Newcastle-Ottawa Quality Assessment Scale for Cohort Studies for randomized controlled trials (RCTs) and non-randomized trials, respectively. Objective response rate (ORR) and progression-free survival (PFS) at 3, 6, and 9 months were investigated as potential early efficacy endpoint candidates for 12-month overall survival (OS), with a strong correlation defined as Pearson's correlation coefficient r ≥ 0.8. RESULTS A total of 64 RCTs comprising 22,725 patients and 106 non-randomized prospective trials involving 10,608 participants were eligible for modeling and external validation, respectively. RCTs examined 15 different cancer types, predominantly non-small-cell lung cancer (NSCLC) (17, 28%), melanoma (9, 14%), and esophageal squamous cell carcinoma (5, 8%). The median sample size of RCTs was 124 patients, and the median follow-up time was 3.2-57.7 months. The ORR (r = 0.38; 95% confidence interval [CI], 0.18-0.54) and PFS (r = 0.42; 95% CI, 0.14-0.64) exhibited weak trial-level correlations with OS. Within ICI treatment arms, the r values of ORR and 3-, 6-, and 9-month PFS with 12-month OS were 0.61 (95% CI, 0.37-0.79), 0.78 (95% CI, 0.62-0.88), 0.84 (95% CI, 0.77-0.90), and 0.86 (95% CI, 0.79-0.90), respectively. External validation of 6-month PFS indicated an acceptable discrepancy between actual and predicted 12-month OS. CONCLUSIONS In non-randomized phase 2 trials on second-line or subsequent ICI therapy in patients with advanced solid cancers, 6-month PFS could serve as an early efficacy endpoint. However, early efficacy endpoints are not recommended in RCTs to replace OS.
Collapse
Affiliation(s)
- Jingqiu Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoding Zhou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Wu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiabao Ma
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Tan
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Songke Wu
- Department of Oncology, People'S Hospital of Cangxi County, Guangyuan, China.
| | - Jie Zhu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| | - Qifeng Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| | - Qiuling Shi
- Center for Cancer Prevention Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| |
Collapse
|
27
|
Liu C, Zou H, Ruan Y, Fang L, Wang B, Cui L, Wu T, Chen Z, Dang T, Lan Y, Zhao W, Zhang C, Meng H, Zhang Y. Multiomics Reveals the Immunologic Features and the Immune Checkpoint Blockade Potential of Colorectal Medullary Carcinoma. Clin Cancer Res 2025; 31:773-786. [PMID: 39651997 PMCID: PMC11831109 DOI: 10.1158/1078-0432.ccr-24-2505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 02/18/2025]
Abstract
PURPOSE Colorectal medullary carcinoma (MeC) is extensive lymphocyte infiltration and is associated with an active immune response. However, studies to comprehensively explore the immune landscape and efficacy of immune checkpoint blockade (ICB) therapy in MeC are limited. EXPERIMENTAL DESIGN We screened 47 cases of MeC from the Harbin Medical University Cancer Hospital cohort. The immunologic characteristics of MeC were analyzed by targeted exon sequencing, NanoString nCounter gene expression sequencing, IHC, multiplexed immunofluorescence, and T-cell antigen receptor sequencing. An additional 47 patients with MeC who received ICB therapy were included in the retrospective analysis to verify the efficacy of immunotherapy. RESULTS Genomically, MeC tends to have a higher proportion of mismatch repair protein deficiency/microsatellite instability (MSI), ARID1A mutation, and ASCL2 amplification. Gene expression shows enriched immune response-related pathways while downregulating oncogenic pathways, such as glycolysis, epithelial-mesenchymal transition, and Wnt/β-catenin signaling. Further immune characterization showed that MeC showed advantages in antigen presentation, co-stimulatory molecules, effector molecules, immune checkpoints, and immune cell abundance. More importantly, both MSI and microsatellite-stable type MeC showed a similar state of high infiltration of immune cells, even better than MSI non-MeC. MeC infiltrated massive highly clonal immune cells, especially intraepithelial CD8+ T cells. In the retrospective cohort, there were 30 patients with MeC who received ICB therapy and achieved complete or partial response with an objective response rate of 63.8%, especially including 16 patients with microsatellite-stable colorectal cancer. CONCLUSIONS MeC is a pathologic subtype with an active immune response and is a promising group for ICB therapy. This heightened immune response was not limited to the patients' microsatellite status.
Collapse
Affiliation(s)
- Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| | - Haoyi Zou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Yuli Ruan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Lin Fang
- Phase I Clinical Research Center, The Affiliated Hospital of Qingdao University in Shandong, Qingdao, China
| | - Bojun Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Luying Cui
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| | - Tong Wu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Zhuo Chen
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Tianjiao Dang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Ya Lan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Wenyuan Zhao
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Chunhui Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| |
Collapse
|
28
|
Wang Y, Hu C, Du T, Li J, Hui K, Jiang X. Combination of potassium oxonate with anti-PD-1 for the treatment of colorectal cancer. Front Oncol 2025; 15:1528004. [PMID: 39990679 PMCID: PMC11842225 DOI: 10.3389/fonc.2025.1528004] [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: 11/14/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction Identification of effective therapies for colorectal cancer (CRC) remains an urgent medical need, especially for the microsatellite stable (MSS) phenotype. In our previous study, potassium oxonate (PO), a uricase inhibitor commonly used for elevating uric acid in mice, unexpectedly showed remarkable inhibition of tumor growth when combined with anti-programmed death-1 (PD-1). Further research demonstrated that the combination of potassium oxonate and anti-PD-1 could reprogram the immune microenvironment. This study aimed to explore the anti-tumor effect of PO combined with anti-PD-1, and investigate the impact on the immunosuppressive tumor microenvironment (TME). Methods We established a syngeneic mouse model of CRC and divided into groups of control group, single drugs group of PO and anti-PD-1, and the combination group. Use the HE staining, immunohistochemistry (IHC) and TUNEL staining of tumor issues to verify the anti-neoplasm of each group. We also tested the changes of TME through flow cytometry of spleen of mice in each group, as well as the IHC of cytokines. Results The co-therapy of PO and anti-PD-1 showed admirable anti-tumor effect compared with the control group and the single drug groups. The TME were tended to an environment beneficial for killing tumors by enhancing chemotactic factor release, increasing CD8+ T cell infiltration and activation, and decreasing the amount of regulatory T cells. Moreover, IFN-γ and IL-2 secretion were found to be enriched in the tumor TME. Conclusion Our study indicated that combination of PO and anti-PD-1 could synergistically suppress CRC progression and altered the tumor microenvironment in favor of antitumor immune responses.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Chenxi Hu
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Tianpeng Du
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiawen Li
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Kaiyuan Hui
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Xiaodong Jiang
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China
| |
Collapse
|
29
|
Luo D, Zhou J, Ruan S, Zhang B, Zhu H, Que Y, Ying S, Li X, Hu Y, Song Z. Overcoming immunotherapy resistance in gastric cancer: insights into mechanisms and emerging strategies. Cell Death Dis 2025; 16:75. [PMID: 39915459 PMCID: PMC11803115 DOI: 10.1038/s41419-025-07385-7] [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: 11/07/2024] [Revised: 01/07/2025] [Accepted: 01/22/2025] [Indexed: 02/09/2025]
Abstract
Gastric cancer (GC) remains a leading cause of cancer-related mortality worldwide, with limited treatment options in advanced stages. Immunotherapy, particularly immune checkpoint inhibitors (ICIs) targeting PD1/PD-L1, has emerged as a promising therapeutic approach. However, a significant proportion of patients exhibit primary or acquired resistance, limiting the overall efficacy of immunotherapy. This review provides a comprehensive analysis of the mechanisms underlying immunotherapy resistance in GC, including the role of the tumor immune microenvironment, dynamic PD-L1 expression, compensatory activation of other immune checkpoints, and tumor genomic instability. Furthermore, the review explores GC-specific factors such as molecular subtypes, unique immune evasion mechanisms, and the impact of Helicobacter pylori infection. We also discuss emerging strategies to overcome resistance, including combination therapies, novel immunotherapeutic approaches, and personalized treatment strategies based on tumor genomics and the immune microenvironment. By highlighting these key areas, this review aims to inform future research directions and clinical practice, ultimately improving outcomes for GC patients undergoing immunotherapy.
Collapse
Affiliation(s)
- Dingtian Luo
- Gastroenterology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jing Zhou
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Shuiliang Ruan
- Gastroenterology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Binzhong Zhang
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Huali Zhu
- Gastroenterology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Yangming Que
- Gastroenterology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Shijie Ying
- Gastroenterology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaowen Li
- Pathology Department, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Yuanmin Hu
- Intensive Care Unit, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Zhengwei Song
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| |
Collapse
|
30
|
Xu L, Xu P, Wang J, Ji H, Zhang L, Tang Z. Advancements in clinical research and emerging therapies for triple-negative breast cancer treatment. Eur J Pharmacol 2025; 988:177202. [PMID: 39675457 DOI: 10.1016/j.ejphar.2024.177202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
Triple-negative breast cancer (TNBC), defined by the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor 2 (HER2) expression, is acknowledged as the most aggressive form of breast cancer (BC), comprising 15%-20% of all primary cases. Despite the prevalence of TNBC, effective and well-tolerated targeted therapies remain limited, with chemotherapy continuing to be the mainstay of treatment. However, the horizon is brightened by recent advancements in immunotherapy and antibody-drug conjugates (ADCs), which have garnered the U.S. Food and Drug Administration (FDA) approval for various stages of TNBC. Poly (ADP-ribose) polymerase inhibitors (PARPi), particularly for TNBC with BRCA mutations, present a promising avenue, albeit with the challenge of resistance that must be addressed. The success of phosphoinositide-3 kinase (PI3K) pathway inhibitors in hormone receptor (HR)-positive BC suggests potential applicability in TNBC, spurring optimism within the research community. This review endeavors to offer a comprehensive synthesis of both established and cutting-edge targeted therapies for TNBC. We delve into the specifics of PARPi, androgen receptor (AR) inhibitors, Cancer stem cells (CSCs), PI3K/Protein Kinase B (AKT)/mammalian target of rapamycin (mTOR), the transforming growth factor-beta (TGF-β), Ntoch, Wnt/β-catenin, hedgehog (Hh) pathway inhibitors, Epigenetic target-mediated drug delivery, ADCs, immune checkpoint inhibitors (ICIs)and novel immunotherapeutic solutions, contextualizing TNBC within current treatment paradigms. By elucidating the mechanisms of these drugs and their prospective clinical applications, we aim to shed light on the challenges and underscore the beacon of hope that translational research and innovative therapies represent for the oncology field.
Collapse
Affiliation(s)
- Lili Xu
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Pengtao Xu
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Jingsong Wang
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, 628000, China
| | - Hui Ji
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Lin Zhang
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China.
| |
Collapse
|
31
|
Guo H, Miao L, Yu C. The efficacy of targeted therapy and/or immunotherapy with or without chemotherapy in patients with colorectal cancer: A network meta-analysis. Eur J Pharmacol 2025; 988:177219. [PMID: 39716565 DOI: 10.1016/j.ejphar.2024.177219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND The use of targeted drugs and immunotherapy has significantly impacted the treatment of Colorectal Cancer. However, horizontal comparison among various regimens is extremely rare. Therefore, we evaluated the survival efficacy of multiple treatment regimens of targeted therapy and/or immunotherapy with or without chemotherapy in patients with Colorectal Cancer. METHODS A systematic search was conducted in PubMed, EMBASE, and Cochrane databases, covering the period from the establishment of the databases to October 29, 2024. To obtain articles that met the inclusion and exclusion criteria and contained the required data for conducting a network meta-analysis (NMA). The NMA evaluated overall survival (OS) and progression-free survival (PFS). RESULTS A total of 90 studies were identified, comprising a sample size of 33,167 subjects. In terms of PFS, compared with simple chemotherapy strategies, most of the other single or combined strategies are significantly effective, among which targeted therapy strategies have more advantages. Encorafenib + Binimetinib + Cetuximab (ENC-BIN-CET) shows significant benefits in all comparisons except when compared with Chemotherapy + Cetuximab + Dalotuzumab (Chemo-CET-DAL), Encorafenib + Cetuximab (ENC-CET), and Panitumumab + Sotorasib (PAN-SOT). The ENC-CET and PAN-SOT targeted strategies also show significant benefits. Pembrolizumab (PEM) monotherapy has advantages over all others except when it is not superior to some targeted strategies. Chemotherapy + Bevacizumab + Atezolizumab is only inferior to some strategies. In terms of OS, the combinations of Chemotherapy + Bevacizumab, ENC-CET, Chemotherapy + Panitumumab, and ENC-BIN-CET are superior to simple chemotherapy regimens. ENC-BIN-CET shows OS benefits in all comparisons except some. ENC-CET significantly improves OS in most cases, and PEM also significantly improves OS in some regimens. In the probability ranking of OS and PFS, ENC-BIN-CET has the best effect, followed by ENC-CET. CONCLUSIONS In conclusion, pembrolizumab is still effective in prolonging survival. Dual- and triple-drug targeted strategies are the best in terms of OS and PFS, and the combination of targeted immunotherapy and chemotherapy also works. However, not all combinations are beneficial. As targeted drugs play an active role, specific drugs for colorectal cancer regimens should be carefully selected.
Collapse
Affiliation(s)
- Haoyan Guo
- Nanhai Hospital of Traditional Chinese Medicine, Jinan University, No.16, Guicheng South Fifth Road, Foshan, Guangdong, 528200, China; Jinan University, Guangzhou, 510632, China
| | - Longjie Miao
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, 518104, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Chengdong Yu
- Nanhai Hospital of Traditional Chinese Medicine, Jinan University, No.16, Guicheng South Fifth Road, Foshan, Guangdong, 528200, China; Jinan University, Guangzhou, 510632, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
32
|
Gao C, Chen L, Zhao L, Su Y, Ma M, Zhang W, Hong X, Xiao L, Xu B, Hu T. Apatinib Degrades PD-L1 and Reconstitutes Colon Cancer Microenvironment via the Regulation of Myoferlin. Cancers (Basel) 2025; 17:524. [PMID: 39941891 PMCID: PMC11816266 DOI: 10.3390/cancers17030524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/20/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND For most colorectal cancer (CRC) patients, expanding the benefits of immunotherapy, particularly through blocking programmed cell death-1 (PD-1) and its ligand (PD-L1), is crucial, especially in cases with limited response to neoadjuvant therapy. This study investigates the role of Myoferlin (MYOF) as a novel target in CRC immunotherapy. METHODS Human CRC cell lines (RKO, HCT116), normal intestinal epithelial cells (HIEC-6), and the murine CRC cell line MC38 were used to study the effects of apatinib and MYOF in CRC cells. RNA sequencing, the CPTAC and TCGA databases, and other molecular and cellular methods were applied to disclose the mechanisms involved. A series of mouse models were established to assess the effects of apatinib and MYOF knockdown on tumor progression, immune cell infiltration, and immune checkpoint protein response. RESULTS We found that MYOF is overexpressed in CRC and linked to immune cell infiltration and checkpoint expression. Suppression of MYOF expression significantly inhibited CRC cell proliferation and migration, as well as reduced PD-L1 protein levels. Integrative analysis showed that apatinib modulates MYOF expression via VEGFR2, resulting in decreased PD-L1 expression, increased CD8+ T cell infiltration, and reduced pro-tumor M2 macrophages. Animal experiments further revealed that apatinib treatment or MYOF knockdown enhanced the efficacy of immune checkpoint blockade (ICB) in CRC. CONCLUSIONS These findings highlight novel antitumor mechanisms of MYOF and suggest that combining apatinib with ICB therapy may improve CRC treatment outcomes, offering a promising strategy to enhance immune responses.
Collapse
Affiliation(s)
- Chunyi Gao
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou 341000, China
| | - Lu Chen
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
| | - Lingying Zhao
- Department of Laboratory Medicine, Shenzhen Children’s Hospital, Shenzhen 518038, China;
| | - Yongcheng Su
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
| | - Miaomiao Ma
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
| | - Wenqing Zhang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
| | - Xiaoting Hong
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
| | - Li Xiao
- Department of Oncology, Zhongshan Hospital of Xiamen University, Xiamen 361004, China;
| | - Beibei Xu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Tianhui Hu
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
33
|
Haldar SD, Kopetz S. Dual checkpoint blockade for microsatellite instability-high colorectal cancer. Lancet 2025; 405:354-356. [PMID: 39874978 DOI: 10.1016/s0140-6736(25)00144-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Affiliation(s)
- S Daniel Haldar
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77584, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77584, USA.
| |
Collapse
|
34
|
Piercey O, Chantrill L, Hsu H, Ma B, Price T, Tan IB, Teng H, Tie J, Desai J. Expert consensus on the optimal management of BRAF V600E-mutant metastatic colorectal cancer in the Asia-Pacific region. Asia Pac J Clin Oncol 2025; 21:31-45. [PMID: 39456063 PMCID: PMC11733838 DOI: 10.1111/ajco.14132] [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: 05/15/2024] [Revised: 09/14/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
The burden of colorectal cancer (CRC) is high in the Asia-Pacific region, and several countries in this region have among the highest and/or fastest growing rates of CRC in the world. A significant proportion of patients will present with or develop metastatic CRC (mCRC), and BRAFV600E-mutant mCRC represents a particularly aggressive phenotype that is less responsive to standard chemotherapies. In light of recent therapeutic advances, an Asia-Pacific expert consensus panel was convened to develop evidence-based recommendations for the diagnosis, treatment, and management of patients with BRAFV600E-mutant mCRC. The expert panel comprised nine medical oncologists from Australia, Hong Kong, Singapore, and Taiwan (the authors), who met to review current literature and develop eight consensus statements that describe the optimal management of BRAFV600E-mutant mCRC in the Asia-Pacific region. As agreed by the expert panel, the consensus statements recommend molecular testing at diagnosis to guide individualized treatment decisions, propose optimal treatment pathways according to microsatellite stability status, advocate for more frequent monitoring of BRAFV600E-mutant mCRC, and discuss local treatment strategies for oligometastatic disease. Together, these expert consensus statements are intended to optimize treatment and improve outcomes for patients with BRAFV600E-mutant mCRC in the Asia-Pacific region.
Collapse
Affiliation(s)
| | - Lorraine Chantrill
- Illawarra Shoalhaven Local Health DistrictIllawarraNew South WalesAustralia
- Faculty of Science, Medicine and HealthUniversity of WollongongWollongongNew South WalesAustralia
| | - Hung‐Chih Hsu
- Division of Hematology OncologyChang Gung Memorial HospitalNew TaipeiTaiwan
- College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Brigette Ma
- State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, Department of Clinical Oncology, Hong Kong Cancer InstituteThe Chinese University of Hong KongHong Kong SARChina
| | - Timothy Price
- The Queen Elizabeth HospitalAdelaideSouth AustraliaAustralia
| | - Iain Beehuat Tan
- Division of Medical OncologyNational Cancer Centre SingaporeSingaporeSingapore
| | - Hao‐Wei Teng
- Department of OncologyTaipei Veterans General HospitalTaipeiTaiwan
| | - Jeanne Tie
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Jayesh Desai
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
35
|
Jiang Y, Liu Y, Huang H, Zhao T, Zhao Z, Gao Y. Effect of RAS mutations and related immune characteristics on the prognosis of patients with MSI-H/dMMR colorectal cancer. Cancer Immunol Immunother 2025; 74:78. [PMID: 39891700 PMCID: PMC11787098 DOI: 10.1007/s00262-024-03926-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/16/2024] [Indexed: 02/03/2025]
Abstract
PURPOSE Microsatellite high instability/deficient mismatch repair (MSI-H/dMMR) colorectal cancer (CRC) has an active tumor microenvironment, rendering it more sensitive to immune checkpoint inhibitors. Given that studies involving patients with MSI-H colorectal cancer with RAS mutations are scarce, we explored the effect of RAS mutations on the TME in patients with MSI-H/dMMR cancer and identified potential prognostic factors. METHODS Seventy-five patients diagnosed with MSI-H/dMMR colorectal cancer were retrospectively enrolled and divided into RAS-mutant and -wild-type groups. The expression levels of CD11c+ dendritic cells, CD4+ T cells, CD8+ T cells, and regulatory T cell (Treg) markers were detected, and prognostic factors were analyzed. RESULTS RAS-mutant MSI-H colorectal patients were more likely to have: (1) higher platelet values; (2) shorter disease-free survival (DFS); (3) lower infiltrated numbers of CD11c+ dendritic cells, CD4+ T lymphocytes, and CD8+ T lymphocytes, and higher infiltrated numbers of Foxp3+ Treg cells. In MSI-H/dMMR CRC patients: (1) the high CD11c + , CD4 +, and CD8 + cells infiltration group had longer DFS than the low-infiltration group, and Foxp3 + cells infiltration was not significantly correlated with DFS; (2) the RAS mutation status, number of CD11c+ cells infiltrated, and carbohydrate antigen 19-9 (CA19-9) level were the potential prognostic factors. CONCLUSION RAS mutations in patients with MSI-H/dMMR CRC may reduce the infiltration of CD11c+ dendritic cells, CD4+ T cells, and CD8+ T cells, and increase the infiltration of Foxp3+ Treg cells to affect the tumor microenvironment of patients. RAS gene status, CD11c + cells infiltration, and CA19-9 level were potential prognostic factors for MSI-H/dMMR CRC.
Collapse
Affiliation(s)
- Yupeng Jiang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yuyao Liu
- Department of Oncology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410004, China
| | - Hong Huang
- Guilin Medical University, Guilin, 541000, China
| | - Tiantian Zhao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Zengyi Zhao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yawen Gao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
36
|
Achalla LSV, Shinde RK, Shukla S, Jogdand SD, Vodithala S. Assessment of HER2/Neu Expression in Colorectal Carcinomas and Its Correlation With Tumor Stage and Histopathology. Cureus 2025; 17:e79721. [PMID: 40161124 PMCID: PMC11954443 DOI: 10.7759/cureus.79721] [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: 01/10/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Colorectal carcinoma (CRC) is the second leading cause of cancer-related mortality worldwide. Within this context, a subset of CRCs shows overexpression of the human epidermal growth factor receptor 2 (HER2/neu), a characteristic also seen in other malignancies like breast and gastric cancers. The success of targeting the HER2 pathway in these cancers has prompted investigations into the potential use of similar interventions in CRC. Aims This research aims to assess HER2/neu expression in CRCs and its correlation with the CRC stage and histopathology, as well as to evaluate the demographic characteristics of CRC patients. Materials and methods This prospective observational study was conducted on a cohort of 40 CRC patients, using histopathology and immunohistochemistry (IHC) sections from the Department of Pathology. Clinical and demographic data were collected over a 24-month period, and routine tissue processing and IHC staining were performed on colectomy tissues. Immunostaining with the HER2/neu marker was conducted for detailed analysis. Data were analyzed using IBM SPSS Statistics for Windows, Version 27.0 (Released 2020; IBM Corp., Armonk, NY, United States). Results Among the 40 CRC cases studied, six cases (15%) exhibited robust membranous positivity, while eight cases (20%) showed moderate focal membranous positivity. Twenty-six cases (65%), however, demonstrated no HER2/neu staining positivity. A significant correlation was found between the histological grade of the tumor and HER2/neu expression (p = 0.0001). Additionally, HER2/neu expression was significantly correlated with lymphovascular invasion (p < 0.0001) and lymph node status (p < 0.047). Conclusion This study found a strong correlation between the tumor's stage, grade, lymph node status, and lymphovascular invasion and HER2/neu expression. Therefore, HER2/neu can be a predictive and therapeutic marker in colorectal cancers. This underscores the potential importance of incorporating these parameters in the clinical evaluation and targeted treatment strategies for CRC patients.
Collapse
Affiliation(s)
- Lakshmi Sai Vijay Achalla
- Department of General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute Of Higher Education and Research, Wardha, IND
| | - Raju K Shinde
- Department of General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute Of Higher Education and Research, Wardha, IND
| | - Samarth Shukla
- Department of Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute Of Higher Education and Research, Wardha, IND
| | - Sangita D Jogdand
- Department of Pharmacology, Jawaharlal Nehru Medical College, Datta Meghe Institute Of Higher Education and Research, Wardha, IND
| | - Sahitya Vodithala
- Department of Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute Of Higher Education and Research, Wardha, IND
| |
Collapse
|
37
|
Das S, Berlin J. Systemic Therapy Improvements Will Render Locoregional Treatments Obsolete for Patients with Cancer with Liver Metastases. Hematol Oncol Clin North Am 2025; 39:191-206. [PMID: 39510673 DOI: 10.1016/j.hoc.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Hepatic metastases are a major cause of morbidity and mortality for patients with cancer. Apart from curative resection, which offers patients the potential for long-term survival, an array of locoregional therapies, with limited evidence of improving survival, are used to treat them. The authors use examples from the realm of gastrointestinal cancer, largely focusing on the experience of patients with neuroendocrine cancer, hepatobiliary cancer, and colorectal cancer, to suggest that current systemic therapies offer, at minimum, similar survival outcomes for patients compared with these locoregional approaches.
Collapse
Affiliation(s)
- Satya Das
- Department of Medicine, Division of Hematology Oncology, Vanderbilt University Medical Center, 777 Preston Research Building, 2220 Pierce Avenue, Nashville, TN 37232, USA.
| | - Jordan Berlin
- Department of Medicine, Division of Hematology Oncology, Vanderbilt University Medical Center, 777 Preston Research Building, 2220 Pierce Avenue, Nashville, TN 37232, USA. https://twitter.com/jordanberlin5
| |
Collapse
|
38
|
Marabelle A, O'Malley DM, Hendifar AE, Ascierto PA, Motola-Kuba D, Penel N, Cassier PA, Bariani G, De Jesus-Acosta A, Doi T, Longo F, Miller WH, Oh DY, Gottfried M, Yao L, Jin F, Gozman A, Maio M. Pembrolizumab in microsatellite-instability-high and mismatch-repair-deficient advanced solid tumors: updated results of the KEYNOTE-158 trial. NATURE CANCER 2025; 6:253-258. [PMID: 39979665 DOI: 10.1038/s43018-024-00894-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/10/2024] [Indexed: 02/22/2025]
Abstract
The phase 2 trial KEYNOTE-158 ( NCT02628067 ) evaluated pembrolizumab in microsatellite-instability-high and mismatch-repair-deficient (MSI-H/dMMR) noncolorectal tumors. With 373 participants (95% with baseline MSI/dMMR documentation) and 4.5 years of follow-up, the primary endpoint of overall response rate was 33.8%. Secondary endpoints of duration of response, overall survival and progression-free survival were 63.2, 19.8 and 4.0 months, respectively. Grade ≥3 treatment-related adverse events occurred in 50 (13%) participants. These results further support pembrolizumab use in MSI-H/dMMR tumors.
Collapse
Affiliation(s)
- Aurelien Marabelle
- Gustave Roussy, Institut National de la Santé et de la Recherche Médicale U1015 & CIC1428, Universite Paris Saclay, Villejuif, France.
| | - David M O'Malley
- The Ohio State University Wexner Medical Center, Columbus, OH, USA
- The James Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | | | - Nicolas Penel
- Centre Oscar Lambret and Lille University, Lille, France
| | | | - Giovanni Bariani
- Instituto do Câncer do Estado de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | - Ana De Jesus-Acosta
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Toshihiko Doi
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Federico Longo
- Hospital Universitario Ramón y Cajal, IRYCIS, CIBERONC, Madrid, Spain
| | - Wilson H Miller
- Jewish General Hospital, Montréal, Québec, Canada
- McGill University, Montréal, Québec, Canada
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | - Lili Yao
- Merck & Co., Inc., Rahway, NJ, USA
| | - Fan Jin
- Merck & Co., Inc., Rahway, NJ, USA
| | | | - Michele Maio
- University of Siena, Siena, Italy
- Center for Immuno-Oncology, Department of Oncology, University Hospital, Siena, Italy
| |
Collapse
|
39
|
Chuang J, Chen Y, Wang J. Narrative review of neoadjuvant therapy in patients with locally advanced colon cancer. Kaohsiung J Med Sci 2025; 41:e12926. [PMID: 39717937 PMCID: PMC11827549 DOI: 10.1002/kjm2.12926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 11/28/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024] Open
Abstract
Colorectal cancer is a leading cause of cancer-related morbidity and mortality worldwide, with more than 1.9 million new cases reported in 2020, and is associated with major survival challenges, particularly in patients with locally advanced colon cancer (LACC). LACC often involves T4 invasion or extensive nodal involvement and requires a multidisciplinary approach for management. Radical surgery followed by adjuvant chemotherapy remains the primary treatment strategy for LACC. However, achieving complete tumor resection (R0) is challenging because locally advanced colon tumors typically infiltrate adjacent organs or nodes. Advancements in LACC treatment have involved neoadjuvant chemotherapy (NACT), neoadjuvant chemoradiotherapy (NACRT), and neoadjuvant immunotherapy (NAIT). Studies such as FOxTROT and PRODIGE 22 have demonstrated that NACT, particularly with FOLFOX or CAPOX, can lead to major tumor downstaging, improved survival rates, and increased R0 resection rates. Predictive biomarkers, such as mismatch repair (MMR) status and T stage, are crucial in identifying candidates who may benefit from NACT. NACRT has demonstrated promise in enhancing tumor regression, particularly in patients with rectal cancer, underscoring its potential for use with LACC. NAIT, particularly for deficient MMR tumors, has emerged as a novel approach, with studies such as NICHE-2 and NICHE-3 reporting excellent pathologic responses and pathologic complete responses. Integrating these therapies can enhance the surgical and survival outcomes of patients with LACC, highlighting the importance of personalized treatment strategies based on tumor characteristics and response to neoadjuvant interventions. This review discusses the evolving landscape of LACC management, focusing on optimizing treatment approaches for improved patient outcomes.
Collapse
Affiliation(s)
- Jen‐Pin Chuang
- Chiayi HospitalMinistry of Health and WelfareChiayiTaiwan
- Department of Surgery, Faculty of Medicine, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- Department of SurgeryNational Cheng Kung University HospitalTainanTaiwan
| | - Yen‐Chen Chen
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University HospitalKaohsiung Medical UniversityKaohsiungTaiwan
- Graduate Institute of Clinical Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Surgery, Faculty of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Jaw‐Yuan Wang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University HospitalKaohsiung Medical UniversityKaohsiungTaiwan
- Graduate Institute of Clinical Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Surgery, Faculty of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Center for Cancer ResearchKaohsiung Medical UniversityKaohsiungTaiwan
| |
Collapse
|
40
|
Jiang SS, Kang ZR, Chen YX, Fang JY. The gut microbiome modulate response to immunotherapy in cancer. SCIENCE CHINA. LIFE SCIENCES 2025; 68:381-396. [PMID: 39235561 DOI: 10.1007/s11427-023-2634-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/05/2024] [Indexed: 09/06/2024]
Abstract
Gut microbiota have been reported to play an important role in the occurrence and development of malignant tumors. Currently, clinical studies have identified specific gut microbiota and its metabolites associated with efficacy of immunotherapy in multiple types of cancers. Preclinical investigations have elucidated that gut microbiota modulate the antitumor immunity and affect the efficacy of cancer immunotherapy. Certain microbiota and its metabolites may favorably remodel the tumor microenvironment by engaging innate and/or adaptive immune cells. Understanding how the gut microbiome interacts with cancer immunotherapy opens new avenues for improving treatment strategies. Fecal microbial transplants, probiotics, dietary interventions, and other strategies targeting the microbiota have shown promise in preclinical studies to enhance the immunotherapy. Ongoing clinical trials are evaluating these approaches. This review presents the recent advancements in understanding the dynamic interplay among the host immunity, the microbiome, and cancer immunotherapy, as well as strategies for modulating the microbiome, with a view to translating into clinical applications.
Collapse
Affiliation(s)
- Shan-Shan Jiang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Zi-Ran Kang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China.
| |
Collapse
|
41
|
He C, Dong W, Lyu Y, Qin Y, Zhong S, Jiang X, Xiao J. Molecular characteristics, clinical significance and cancer‑immune interactions of pyroptosis‑related genes in colorectal cancer. Oncol Lett 2025; 29:89. [PMID: 39677414 PMCID: PMC11638898 DOI: 10.3892/ol.2024.14835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/28/2024] [Indexed: 12/17/2024] Open
Abstract
Colorectal cancer (CRC) is a malignant tumor with poor prognosis. Pyroptosis is a newly discovered type of programmed cell death that is typically accompanied by a strong inflammatory response. Accumulating evidence suggests that pyroptosis-related genes (PRGs) may have important roles in the development of malignant tumors. However, the association between PRG expression and clinical outcomes in CRC remain unclear. In the present study, the genetic variations and transcriptional patterns of 52 PRGs were comprehensively analyzed using cohorts from The Cancer Genome Atlas and Gene Expression Omnibus and the mRNA expression levels of 7 PRGs in collected CRC samples were validated using reverse transcription-quantitative PCR. Using LASSO-Cox analysis, a PRG score was then generated and the relationship between the PRG score and prognosis, immune cell infiltration and drug sensitivity in CRC was uncovered. In the present study, the mutation and expression patterns of PRGs were analyzed and it was found that these genes were differentially expressed in CRC tissues compared with normal tissues. Based on the expression patterns of the PRGs, patients with CRC were divided into two subtypes (cluster A and B), of which cluster B had an improved prognosis and a higher abundance of immune cells. Next, differentially expressed genes between clusters A and B were identified and a PRG risk score closely related to the prognosis of CRC was constructed. Then, a nomogram for evaluating the overall survival of patients was constructed. Furthermore, a low PRG risk score was characterized by immune activation and closely related to the microsatellite instability-high pattern. Additionally, the PRG risk score was notably correlated with drug sensitivity. In conclusion, the mutation and expression characteristics of PRGs in CRC were comprehensively analyzed and a prognostic PRG signature was constructed in the present study. This signature may predict immune cell infiltration and therapeutic response in CRC, providing new insights into the prognosis and treatment of CRC.
Collapse
Affiliation(s)
- Chenglong He
- Department of Oncology, Zhongshan City People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Wenjing Dong
- Department of Oncology, Zhongshan City People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Yanhua Lyu
- Department of Oncology, Zhongshan City People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Yan Qin
- Department of Oncology, Zhongshan City People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Siquan Zhong
- Department of Oncology, Zhongshan City People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Xiaomei Jiang
- Department of Oncology, Zhongshan City People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Jianjun Xiao
- Department of Oncology, Zhongshan City People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| |
Collapse
|
42
|
André T, Elez E, Lenz HJ, Jensen LH, Touchefeu Y, Van Cutsem E, Garcia-Carbonero R, Tougeron D, Mendez GA, Schenker M, de la Fouchardiere C, Limon ML, Yoshino T, Li J, Manzano Mozo JL, Dahan L, Tortora G, Chalabi M, Goekkurt E, Braghiroli MI, Joshi R, Cil T, Aubin F, Cela E, Chen T, Lei M, Jin L, Blum SI, Lonardi S. Nivolumab plus ipilimumab versus nivolumab in microsatellite instability-high metastatic colorectal cancer (CheckMate 8HW): a randomised, open-label, phase 3 trial. Lancet 2025; 405:383-395. [PMID: 39874977 DOI: 10.1016/s0140-6736(24)02848-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/30/2025]
Abstract
BACKGROUND CheckMate 8HW prespecified dual primary endpoints, assessed in patients with centrally confirmed microsatellite instability-high or mismatch repair-deficient status: progression-free survival with nivolumab plus ipilimumab compared with chemotherapy as first-line therapy and progression-free survival with nivolumab plus ipilimumab compared with nivolumab alone, regardless of previous systemic treatment for metastatic disease. In our previous report, nivolumab plus ipilimumab showed superior progression-free survival versus chemotherapy in first-line microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer in the CheckMate 8HW trial. Here, we report results from the prespecified interim analysis for the other primary endpoint of progression-free survival for nivolumab plus ipilimumab versus nivolumab across all treatment lines. METHODS CheckMate 8HW is a randomised, open-label, international, phase 3 trial at 128 hospitals and cancer centres across 23 countries. Immunotherapy-naive adults with unresectable or metastatic colorectal cancer across different lines of therapy and microsatellite instability-high or mismatch repair-deficient status per local testing were randomly assigned (2:2:1) to nivolumab plus ipilimumab (nivolumab 240 mg, ipilimumab 1 mg/kg, every 3 weeks for four doses; then nivolumab 480 mg every 4 weeks; all intravenously), nivolumab (240 mg every 2 weeks for six doses, then 480 mg every 4 weeks; all intravenously), or chemotherapy with or without targeted therapies. The dual independent primary endpoints were progression-free survival by blinded independent central review with nivolumab plus ipilimumab versus chemotherapy (first line) and progression-free survival by blinded independent central review with nivolumab plus ipilimumab versus nivolumab (all lines) in patients with centrally confirmed microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer. This study is registered with ClinicalTrials.gov (NCT04008030). FINDINGS Between Aug 16, 2019, and April 10, 2023, 707 patients were randomly assigned to nivolumab plus ipilimumab (n=354) or nivolumab alone (n=353). 296 (84%) of 354 patients in the nivolumab plus ipilimumab group and 286 (81%) of 353 patients in the nivolumab group were centrally confirmed to have microsatellite instability-high or mismatch repair-deficient status. At the data cutoff on Aug 28, 2024, median follow-up (from randomisation to data cutoff) was 47·0 months (IQR 38·4 to 53·2). Nivolumab plus ipilimumab treatment showed significant and clinically meaningful improvement in progression-free survival versus nivolumab (hazard ratio 0·62, 95% CI 0·48-0·81; p=0·0003). Median progression-free survival was not reached with nivolumab plus ipilimumab (95% CI 53·8 to not estimable) and was 39·3 months with nivolumab (22·1 to not estimable). Treatment-related adverse events of any grade occurred in 285 (81%) of 352 patients receiving nivolumab plus ipilimumab and in 249 (71%) of 351 patients receiving nivolumab; grade 3 or 4 treatment-related adverse events occurred in 78 (22%) and 50 (14%) patients, respectively. There were three treatment-related deaths: one event of myocarditis and pneumonitis each in the nivolumab plus ipilimumab group and one pneumonitis event in the nivolumab group. INTERPRETATION Nivolumab plus ipilimumab showed superior progression-free survival versus nivolumab across all treatment lines, with a manageable safety profile, in patients with microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer. These results, together with the first-line results of superior progression-free survival with nivolumab plus ipilimumab versus chemotherapy, suggest nivolumab plus ipilimumab as a potential new standard of care for patients with microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer. FUNDING Bristol Myers Squibb and Ono Pharmaceutical.
Collapse
Affiliation(s)
- Thierry André
- Sorbonne Université, Hôpital Saint Antoine, Assistance Publique Hôpitaux de Paris, Paris, France; Unité Mixte de Recherche Scientifique 938, SIRIC CURAMUS, Paris, France.
| | - Elena Elez
- Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Heinz-Josef Lenz
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | - Yann Touchefeu
- Institut des Maladies de l'Appareil Digestif, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg and University of Leuven, Leuven, Belgium
| | | | - David Tougeron
- Centre Hospitalier Universitaire de Poitiers Site de la Miletrie, Poitiers, France
| | | | - Michael Schenker
- Centrul de Oncologie Sf Nectarie, Craiova, Romania; University of Medicine and Pharmacy, Craiova, Romania
| | | | | | | | - Jin Li
- Shanghai East Hospital, Shanghai, China
| | - Jose Luis Manzano Mozo
- Institut Català d'Oncologia, Hospital Universitario Germans Trias i Pujol, Badalona, Spain
| | | | | | | | - Eray Goekkurt
- Hematology-Oncology Practice Eppendorf and University Cancer Center Hamburg, Hamburg, Germany
| | | | | | - Timucin Cil
- University of Health Sciences, Adana Faculty of Medicine, Adana City Education and Research Hospital, Adana, Turkey
| | - Francine Aubin
- Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Elvis Cela
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Tian Chen
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Ming Lei
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Lixian Jin
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | - Sara Lonardi
- Istituto Oncologico Veneto IOV-IRCCS, Padua, Italy
| |
Collapse
|
43
|
Jin Y, Liao L, Chen Q, Tang B, Jiang J, Zhu J, Bai M. Multi-omics analysis reveals that neutrophil extracellular traps related gene TIMP1 promotes CRC progression and influences ferroptosis. Cancer Cell Int 2025; 25:31. [PMID: 39891145 PMCID: PMC11786501 DOI: 10.1186/s12935-025-03643-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: 05/09/2024] [Accepted: 01/08/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Previous studies have found that neutrophil extracellular traps (NETs) are highly expressed in colorectal cancer (CRC) and are associated with poor prognosis. Currently, there are few studies on the relationship between NETs and CRC, so we tried to explore new markers based on NETs to assist in the treatment of CRC. METHOD We jointly screened three major NETs genes through machine learning. Large-sample RNA transcriptome and single-cell transcriptome analysis further confirmed that TIMP1 is a core gene in NETs. We used small interfering RNA to knockdown TIMP1, and verified the ability of TIMP1 in CRC proliferation, invasion and migration through western blot, transwell, cell scratch assay, cell clone formation and other experiments. RESULT We screened out three major NETs Genes: TIMP1, F3, and CRISPLD2 based on machine learning. The NETs score constructed based on this not only predicts the prognosis of CRC patients but also shows significant differences in MSI status, chenckpoints expression, and predicted efficacy of PD-L1 targeted therapy. Transcriptome and single-cell data reveal that TIMP1 is highly expressed in neutrophils and is associated with poor prognosis in colorectal cancer patients and the occurrence of ferroptosis. Biological experiments have proven that TIMP1 can promote the proliferation, invasion and migration of CRC. CONCLUDE Bioinformatics analysis combined with experimental verification showed that TIMP1 is related to ferroptosis and plays a promoting role in the invasion, migration and proliferation of CRC.
Collapse
Affiliation(s)
- Yuzhao Jin
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, Hangzhou, 310000, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China
| | - Luyu Liao
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, Hangzhou, 310000, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China
| | - Qianping Chen
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, Hangzhou, 310000, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China
| | - Bufu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jin Jiang
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, 31400, China
| | - Ji Zhu
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China.
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, Hangzhou, 310000, China.
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China.
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China.
| | - Minghua Bai
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China.
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou, Hangzhou, 310000, China.
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310000, China.
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China.
| |
Collapse
|
44
|
Negro S, Perissinotto E, Mammi I, Crivellari G, Schiavi F, Cappello F, Spolverato G, Ferrari D, Rausa E, Vitellaro M, Fassan M, Cavestro GM, Mannucci A, Lonardi S, Bergamo F, Urso EDL. Emerging therapeutic strategies in Lynch syndrome-associated colorectal cancer and the role of MMR testing. TUMORI JOURNAL 2025:3008916241310706. [PMID: 39882759 DOI: 10.1177/03008916241310706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Lynch syndrome is the most common hereditary cancer predisposition, accounting for 1-5% of colorectal cancer cases, and is driven by germline mutations in DNA mismatch repair genes. Despite established diagnostic criteria, such as the Amsterdam guidelines, Lynch syndrome remains largely underdiagnosed. To address this gap, universal tumour screening has been introduced for all newly diagnosed cases of colorectal cancer and endometrial cancer, significantly improving early detection. The surgical management of colorectal cancer in patients with Lynch syndrome remains controversial. While extended colectomy reduces the risk of metachronous colorectal cancer, surgical strategies must be carefully individualised based on patient-specific factors. Chemoprevention with aspirin has shown promise in reducing the risk of colorectal cancer, with ongoing trials investigating optimal dosing. Immunotherapy, particularly immune checkpoint inhibitors, has revolutionised the treatment of Microsatellite Instability-High/deficient Mismatch Repair colorectal cancer, offering durable responses and significant survival benefits. In addition, the neoadjuvant use of immune checkpoint inhibitors is paving the way for non-surgical interventions, potentially transforming the management of colorectal cancer in patients with Lynch syndrome. A multidisciplinary approach and continued research are essential to optimise cancer prevention, treatment and quality of life for people with Lynch syndrome.
Collapse
Affiliation(s)
- Silvia Negro
- 3rd Surgical Unit, Department of Surgical, Gastroenterological and Oncological Sciences, University of Padua, Padua, Italy
| | - Eleonora Perissinotto
- Medical Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Isabella Mammi
- Unità Tumori Ereditari, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Gino Crivellari
- Unità Tumori Ereditari, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Francesca Schiavi
- Unità Tumori Ereditari, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Filippo Cappello
- Pathological Anatomy Unit, University Hospital of Padova, Padova, Italy
| | - Gaya Spolverato
- 3rd Surgical Unit, Department of Surgical, Gastroenterological and Oncological Sciences, University of Padua, Padua, Italy
| | - Davide Ferrari
- Department of Surgery, IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Emanuele Rausa
- Department of Surgery, IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco Vitellaro
- Department of Surgery, IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy; Department of Pathology, Azienda ULSS2 Marca Trevigiana, Treviso
| | - Giulia Martina Cavestro
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Mannucci
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Lonardi
- Medical Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Francesca Bergamo
- Medical Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Emanuele D L Urso
- 3rd Surgical Unit, Department of Surgical, Gastroenterological and Oncological Sciences, University of Padua, Padua, Italy
| |
Collapse
|
45
|
Zhang D, Chen T. The Efficacy and Safety of Durvalumab and Tremelimumab with Concomitant Treatment for MSS/pMMR Metastatic Colorectal Cancer: A Single Arm Meta-Analysis. J Gastrointest Cancer 2025; 56:56. [PMID: 39875748 DOI: 10.1007/s12029-025-01181-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2025] [Indexed: 01/30/2025]
Abstract
OBJECTIVES To address the issue that most microsatellite-stable (MSS) and proficient mismatch repair (pMMR) metastatic colorectal cancer (mCRC) patients have minimal response to immunotherapy, this meta-analysis evaluated the efficacy and safety of durvalumab and tremelimumab with concomitant treatment in treating MSS/pMMR metastatic colorectal cancer. METHODS All included trials were prospective studies with a median patient age of 63 years, of which 94.2% were MSS/pMMR mCRC patients, with a male to female ratio of 1.5:1. Based on durvalumab and tremelimumab treatment, one study performed surgical resection on resectable cases, while the other four studies performed radiotherapy or chemotherapy on unresectable cases. Analyses include objective response rate (ORR).etc. for drug activity, overall survival (OS) and progression-free survival (PFS) for therapeutic efficacy, and adverse events (AEs) for safety. The risk of bias was assessed by sensitivity analysis. RESULTS 5 studies involving 228 patients were included in this meta-analysis. The pooled estimates showed a median OS of 9.26 months, median PFS of 2.53 months, partial response (PR) of 13.6%, stable disease (SD) of 32.8%, ORR of 12.5% and disease control rate (DCR) of 65.4%. AEs were generally low, with pruritus (27.5%), diarrhea (28.8%), and fatigue (53.9%) being the most common, while other AEs occurred at less frequencies. CONCLUSIONS Durvalumab and tremelimumab with concomitant treatment for MSS/pMMR mCRC patients is relatively effective and safe, which is helpful in addressing the problem of mCRC with MSS/pMMR that has minimal response to immunotherapy.
Collapse
Affiliation(s)
- Danning Zhang
- College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.
- Sichuan University, Xinhangang Street, Shuangliu District, Chengdu, 610000, Sichuan, China.
| | - Tianyu Chen
- Computer Science, Changchun University of Science and Technology, Changchun, 130022, Jilin, China
| |
Collapse
|
46
|
Zhang H, Huang J, Xu H, Yin N, Zhou L, Xue J, Ren M. Neoadjuvant immunotherapy for DNA mismatch repair proficient/microsatellite stable non-metastatic rectal cancer: a systematic review and meta-analysis. Front Immunol 2025; 16:1523455. [PMID: 39931055 PMCID: PMC11808008 DOI: 10.3389/fimmu.2025.1523455] [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: 11/06/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
Background Neoadjuvant immunotherapy (NIT) has been endorsed by clinical guidelines for the management of DNA mismatch repair deficiency/microsatellite instability-high (dMMR/MSI-H) locally advanced rectal cancer (LARC). Nonetheless, the therapeutic efficacy of NIT in mismatch repair-proficient/microsatellite stable (pMMR/MSS) non-metastatic rectal cancer (RC) remain pending matters. Therefore, a meta-analysis was carried out to assess the efficacy and safety of NIT in patients with non-metastatic pMMR/MSS RC. Methods PubMed, Embase, Web of Science, the Cochrane Library, ClinicalTrials.gov, ASCO and ESMO were searched to obtain related studies up to July 2024. Two reviewers independently screened the included articles and extracted the pertinent data. The risk of publication bias was assessed by Begg or Egger tests and in cases of publication bias, the trim and fill method was applied. Heterogeneity was assessed using I 2 statistics. Results Thirteen articles including 582 eligible patients were analyzed. The pooled pCR, MPR, cCR and anus preservation rate were 37%, 57%, 26% and 77% separately and the incidence of irAEs≥3 grades and TRAEs≥3 grades were 3% and 29%, respectively. Non-metastatic pMMR/MSS RC receiving the short-course radiotherapy (SCRT) in neoadjuvant setting exhibited superior pooled pCR and MPR than long-course radiotherapy (LCRT) without upregulating the incidence of adverse effects. Furthermore, patients with MSS RC underwent neoadjuvant treatment with anti-PD-1 inhibitors demonstrated higher pooled pCR, MPR, cCR compared to those receiving PD-L1 inhibitors. Additionally, yielded improved pooled MPR and anal preservation rates compared to sequential immuno-radiotherapy (63.4% vs 51.2% and 88.5% vs 69.9%), without raising the incidence of irAEs≥3 grade. Interestingly, RC patients with lymph node metastasis showed a higher pooled pCR than those without lymph node metastasis (43% vs 35%). Conclusion NIT was linked to favorable response rates and anal preservation, alongside an acceptable safety profile. Non-metastatic pMMR/MSS RC patients receiving SCRT, PD-1 inhibitors, or concurrent immuno-radiotherapy in the neoadjuvant setting exhibited enhanced outcomes. This meta-analysis provides evidence for further exploration and application of NIT in non-metastatic pMMR/MSS RC and highlights the potential for organ preservation with this approach. The relatively small sample size and the uneven quality of included studies may have had some impact on the generality of the results. Therefore, further analysis with a higher number of high-quality studies is needed to verify the conclusions. Systematic review registration https://inplasy.com/, identifier: INPLASY202470110.
Collapse
Affiliation(s)
- Huan Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing Huang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huanji Xu
- Abdominal Oncology Ward, Division of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Nanhao Yin
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liyan Zhou
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Ren
- Abdominal Oncology Ward, Division of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
47
|
Torounidou N, Yerolatsite M, Zarkavelis G, Amylidi AL, Karafousia V, Kampletsas E, Mauri D. Treatment sequencing in metastatic colorectal cancer. Contemp Oncol (Pozn) 2025; 28:283-290. [PMID: 39935759 PMCID: PMC11809563 DOI: 10.5114/wo.2024.146982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/16/2024] [Indexed: 02/13/2025] Open
Abstract
Metastatic colorectal cancer (mCRC) remains a significant challenge in contemporary oncology, with treatment sequencing playing a pivotal role in reported patient outcomes. Although currently available therapeutic options have led to improved median overall survival rates, the prognosis for patients with stage IV disease still remains dismal. This article provides a comprehensive review of the current landscape of treatment sequencing strategies in mCRC, focusing on the rationale behind various approaches and the evolving evidence supporting their efficacy. We consider the roles of chemotherapy, targeted therapy, and immunotherapy agents, as well as emerging trends in personalized medicine and sequential therapy regimens. The concept of a "continuum of care" has emerged as a fundamental principle, aiming to provide all available therapies through an individualized approach to maximize clinical benefit. By elucidating the underlying complexities of treatment sequencing in mCRC, this review aims to guide clinicians in optimizing therapeutic strategies and improving patient care.
Collapse
Affiliation(s)
- Nanteznta Torounidou
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Melina Yerolatsite
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - George Zarkavelis
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | - Anna-Lea Amylidi
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Vaia Karafousia
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
| | - Eleftherios Kampletsas
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | - Davide Mauri
- Department of Medical Oncology, University Hospital of Ioannina, Ioannina, Greece
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| |
Collapse
|
48
|
Li J, Liu J, Yang S, Xia Y, Meng Q, Sun B, Liu Y, Zhao B, Jin J, Xu H, Wang L, Zhang P, Cheng Z. PD-L1 positive platelets mediate resistance to immune checkpoint inhibitors in patients with colorectal cancer. Cell Commun Signal 2025; 23:29. [PMID: 39815258 PMCID: PMC11737274 DOI: 10.1186/s12964-025-02034-0] [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: 07/12/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Programmed cell death ligand 1 (PD-L1) expression on immune cells is correlated with the efficacy of immune checkpoint inhibitor (ICI) therapy in various types of cancer. Platelets are important components of the tumour microenvironment (TME) and are widely involved in the development of many types of cancer including colorectal cancer (CRC). However, the role of PD-L1 positive platelets in ICI therapy for CRC remains unknown. We hypothesized that PD-L1 positive platelets trigger and sustain CRC immunosuppression. METHODS The functional depletion effects of PD-L1 positive platelets on TME and immune cells were measured via western blotting, immunofluorescence staining, qRT-PCR, ELISpot and flow cytometry. In vivo, CD274 knockout (KO), CD8a KO, platelet-specific KO (PF4-Cre-Hsp90b1flox/flox) mouse models and a subcutaneous tumour model treated with aspirin and PD-L1 mAb were established in C57BL/6 N mice. RESULTS We found that PD-L1 positive platelets are correlated with a poor prognosis, CD8 + T cell exhaustion and serve as a novel noninvasive biomarker for predicting immunotherapy efficacy in patients with CRC. The transfer of PD-L1 from tumour cells to platelets in the TME depends on direct cell contact via the fibronectin-1/GPIbα/integrin α5β1 pathway. In turn, platelets can also induce PD-L1 expression on cancer cells. Animal experiments revealed that antiplatelet pharmacological agents and genetic knockout of platelets potentiated the antitumour effect of the PD-L1 mAb treatment in a CD8 + T cell dependent manner. CONCLUSIONS Our data suggest that PD-L1 positive platelets suppress CD8 + T cell immunity. Clinical combination treatment with ICIs and antiplatelet agents may be an effective therapeutic strategy for treating CRC.
Collapse
Affiliation(s)
- Jiacheng Li
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Jia Liu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Shifeng Yang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Yu Xia
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Qingzhe Meng
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Biying Sun
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Yansong Liu
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Bin Zhao
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Jiaqi Jin
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Hui Xu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China.
| | - Lihong Wang
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China.
| | - Pengxia Zhang
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China.
| | - Zhuoxin Cheng
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China.
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China.
| |
Collapse
|
49
|
Yuan M, Zheng Y, Wang F, Bai N, Zhang H, Bian Y, Liu H, He X. Discussion on the optimization of personalized medication using information systems based on pharmacogenomics: an example using colorectal cancer. Front Pharmacol 2025; 15:1516469. [PMID: 39877392 PMCID: PMC11772163 DOI: 10.3389/fphar.2024.1516469] [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: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025] Open
Abstract
Pharmacogenomics (PGx) is a powerful tool for clinical optimization of drug efficacy and safety. However, due to many factors affecting drugs in the real world, PGx still accounts for a small proportion of actual clinical application scenarios. Therefore, based on the information software, pharmacists use their professional advantages to integrate PGx into all aspects of pharmaceutical care, which is conducive to promoting the development of personalized medicine. In this paper, the establishment of an information software platform is summarized for the optimization of a personalized medication program based on PGx. Taking colorectal cancers (CRC) as an example, this paper also discusses the role of PGx in different working modes and participation in drug management of CRC patients by pharmacists with the help of information systems. Finally, we summarized the recommendations of different PGx guidelines to provide reference for the follow-up personalized pharmaceutical care.
Collapse
Affiliation(s)
- Mengying Yuan
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuankun Zheng
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fei Wang
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Niuniu Bai
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Haoling Zhang
- Department of Pharmacy, Yuncheng Central Hospital, Yuncheng, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hao Liu
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xia He
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
50
|
Wang Q, Yu M, Zhang S. The characteristics of the tumor immune microenvironment in colorectal cancer with different MSI status and current therapeutic strategies. Front Immunol 2025; 15:1440830. [PMID: 39877377 PMCID: PMC11772360 DOI: 10.3389/fimmu.2024.1440830] [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: 05/30/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Colorectal cancer (CRC) remains a significant cause of cancer-related mortality worldwide. Despite advancements in surgery, chemotherapy, and radiotherapy, the effectiveness of these conventional treatments is limited, particularly in advanced cases. Therefore, transition to novel treatment is urgently needed. Immunotherapy, especially immune checkpoint inhibitors (ICIs), has shown promise in improving outcomes for CRC patients. Notably, patients with deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) tumors often benefit from ICIs, while the majority of CRC cases, which exhibit proficient mismatch repair (pMMR) or microsatellite-stable (MSS) status, generally show resistance to this approach. It is assumed that the MSI phenotype cause some changes in the tumor microenvironment (TME), thus triggering antitumor immunity and leading to response to immunotherapy. Understanding these differences in the TME relative to MSI status is essential for developing more effective therapeutic strategies. This review provides an overview of the TME components in CRC and explores current approaches aimed at enhancing ICI efficacy in MSS CRC.
Collapse
Affiliation(s)
- Qingzhe Wang
- Department of Targeting Therapy and Immunology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Yu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shuang Zhang
- Department of Targeting Therapy and Immunology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|