Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Aug 24, 2024; 15(8): 968-974
Published online Aug 24, 2024. doi: 10.5306/wjco.v15.i8.968
Pyroptosis: A promising biomarker for predicting colorectal cancer prognosis and enhancing immunotherapy efficacy
Jia-Yi Wang, Yu-Hao Lu, Fang Li, Mo-Li Huang, School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou 215123, Jiangsu Province, China
ORCID number: Mo-Li Huang (0000-0001-5543-9334).
Author contributions: Wang JY and Lu YH contributed to manuscript writing; Huang ML designed the overall concept and outline of the manuscript; Li F and Huang ML contributed to manuscript revision; all authors approved the final manuscript.
Supported by National Natural Science Foundation of China, No. 32370598 and No. 31971117.
Conflict-of-interest statement: The authors have no conflicts of interest to disclose.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Mo-Li Huang, PhD, Professor, School of Biology and Basic Medical Sciences, Medical College of Soochow University, No. 199 Renai Road, Suzhou 215123, Jiangsu Province, China. huangml@suda.edu.cn
Received: March 14, 2024
Revised: May 24, 2024
Accepted: July 2, 2024
Published online: August 24, 2024
Processing time: 155 Days and 4.8 Hours

Abstract

In this editorial, we comment on the article by Zhu et al published in the recent issue of the World Journal of Clinical Oncology. We focus specifically on the characteristics and mechanisms of pyroptosis and the impact of changes in the tumor immune microenvironment (TIME) on cancer prognosis. Pyroptosis is a distinct form of programmed cell death; its occurrence can change the TIME and regulate the growth and spread of tumors and therefore is significantly correlated with cancer prognosis. Previous research has demonstrated that pyroptosis-related genes can be used in prognostic models for various types of cancer. These models enhance the mechanistic understanding of tumor evolution and serve as valuable guides for clinical treatment decision-making. Nevertheless, further studies are required to thoroughly understand the function of pyroptosis within the TIME and to assess its mode of action. Such studies should reveal new tumor therapeutic targets and more successful tumor immunotherapy strategies.

Key Words: Pyroptosis; Colorectal cancer liver metastases; Organoid model; Immunotherapy; Prognostic biomarker

Core Tip: Pyroptosis plays a vital role in tumor immunotherapy by triggering robust inflammatory responses and significantly inhibiting tumors. Pyroptosis results in the release of copious amounts of inflammatory cytokines and tumor-associated antigens that stimulate antigen-presenting cells, thereby initiating adaptive immune responses. Therefore, inducing pyroptosis is a promising novel immunotherapy approach for tumors.



INTRODUCTION

Pyroptosis is a form of programmed cell death that was initially misinterpreted as apoptosis due to their similar characteristics, such as caspase dependency, DNA damage, and nuclear condensation[1,2]. However, pyroptosis is a unique inflammatory programmed cell death process distinct from apoptosis[3-10] (Table 1). The term pyroptosis was first introduced in 2001 and originated from the Greek words “pyro” and “ptosis”, which mean “fire” and “falling”, respectively[11]. With the exception of DFNB59, which lacks pore-forming capacity, gasdermin protein family members are responsible for mediating pyroptosis[12-14]. This protein family has two conserved domains: The N-terminal pore-forming domain (PFD) and the C-terminal repressor domain (RD)[15]. At rest, gasdermin proteins preserve their oligomeric structure via PFD-RD interactions, with the latter curbing the cytotoxic potential of the former. Upon cellular exposure to external or internal stimuli, these proteins are cleaved by specific caspases (caspase-1/4/5/11) or granzymes[16-20]. During this process, the N-terminal PFD dissociates from the C-terminal RD, leading to the oligomerization of PFD. This event results in the formation of pores on the cell membrane, thereby enabling the release of inflammatory molecules and inducing cell swelling, culminating in pyroptosis[21].

Table 1 Differences between pyroptosis and apoptosis.

Apoptosis[5-9]
Pyroptosis[1,2,19-21]
DefinitionThe orderly death of cells autonomously controlled by genes in order to maintain a stable internal environmentAn inflammatory programmed cell death triggered by various pathological stimuli, such as stroke, heart attack, cancer, and microbial infection
Trigger factorGene regulation under physiological conditionsPathological stimulation
Cell morphological changesThe cell size is usually reduced, the cell membrane structure is intact, and the organelles are intactThe cell size is usually enlarged and cell morphology is deformed, the cell membrane is broken, and the organelles are deformed
Marker eventsNucleoprotein, cytoskeleton, and protein crosslinking are destroyed, phagocyte ligands are expressed, and apoptotic bodies are formedThe nucleus is concentrated and pyrosome is formed
DNA changeDNA is degraded into fragments of 180-200 bp and its integral multiplesDNA is randomly degraded
Molecular mechanismCaspase-2/3/6/7/8/9/10, in which caspase-3 is the main regulator of apoptosisHuman caspase-1/4/5; murine caspase-1/11
Detection indexesEarly apoptosis detection indicators: Annexin V and JC-1; metaphase apoptosis detection index: Caspase; late apoptotic detection index: TUNEL stainingNo specific detection indicators, but the cell morphology could be observed by scanning electron microscopy and TUNEL staining, GSDMD immunofluorescence staining could be used to detect the expression levels of pyro related genes or proteins (caspase-1/4/5/11, etc.), and the levels of inflammatory factors (IL-1β, IL-18, etc.) could be detected by ELISA. CCK-8 assay can be used to determine cell viability
DependencyThe endogenous pathway (also called the mitochondrial pathway) is based on caspase-9Classical pathway: Based on caspase-1; nonclassical pathway: Based on caspase-4/5/11

Extensive scholarly research has revealed a strong correlation between pyroptosis and the onset and spread of numerous cancers[22-24]. Chronic inflammatory infiltration increases the risk of cancer. In particular, pyroptosis-induced release of cytokines such as IL-1 and IL-18 might increase the risk of carcinogenesis and metastasis[25]. Nevertheless, Li et al[26] reported that pyroptosis impedes tumor progression through the induction of tumor cell death and activation of the immune system. The concurrent release of cytokines during pyroptosis not only spurs the activation of various immune cells but also modulates the tumor immune microenvironment (TIME) by interacting with key immune checkpoints such as PD-1/PD-L1, indicating promising implications for tumor immunotherapy. The intricate mechanisms by which pyroptosis can stimulate an immune response while simultaneously mitigating its tumor-promoting effects remain a subject for further research. However, current evidence indicates that the manifestations of pyroptosis are closely associated with the onset and progression of colorectal cancer (CRC) and other types of tumors, underscoring the significant potential of these biological markers for evaluating therapeutic outcomes and predicting patient prognoses in the field of oncology. For instance, Dihlmann et al[27] reported that AIM2 inflammasome-mediated pyroptosis is the underlying mechanism of CRC progression. Another study revealed that gasdermin C, an oncogene that is not present in normal colorectal tissues, is upregulated due to inactivating mutations in TGFBR2 in CRC tissues[28].

In summary, pyroptosis triggers inflammation that activates macrophages and boosts T-cell-mediated antitumor immunity, thereby regulating the TIME to effectively suppress tumor growth and spread (Figure 1)[26,29]. Current research focuses on inhibiting tumor progression through the regulation of pyroptosis and its combination with chemotherapy and immunotherapy. In addition, pyroptosis biomarkers are significant for predicting the efficacy and prognosis of CRC and other cancers and could serve as potential therapeutic targets.

Figure 1
Figure 1 Role of pyroptosis in regulating the tumor immune microenvironment and tumor growth. In the canonical pathway, inflammasomes recognize exogenous pathogens and endogenous damage. The intracellular sensor proteins NLRP1, NLRP3, NLRC4, AIM2, Pyrin, and NLRC4 recruit ASC and then activate caspase-1. In the noncanonical pathway, lipopolysaccharide can activate caspase-4/5/11. Activated caspase-1/4/5/11 can cleave GSDMD to release N-GSDMD and pro-IL-1β/18, promoting their maturation. On one hand, N-GSDMD can form nonselective pores, further causing water influx, lysis, and death. Additionally, IL-1β and IL-18 can be released via these pores. On the other hand, N-GSDM, IL-1β, and IL-18 can affect immune cells such as cancer cells in the tumor immune microenvironment, tumor-associated macrophages, CD8+ T cells, and NK cells. Therefore, pyroptosis is a complex regulatory network that acts as a bridge between the immune system and tumors and as a double-edged sword in the tumor immune microenvironment. In this way, it influences tumorigenesis, leading to protumor effects and antitumor effects. LPS: Lipopolysaccharide; TIME: Tumor immune microenvironment.
INFLUENCE OF THE TIME IN PROMOTING IMMUNE EVASION, TUMOR PROGRESSION, AND IMMUNOTHERAPY

The TIME encompasses cellular and acellular components, including cancer cells and immune cells such as T cells, B cells, NK cells, dendritic cells, myeloid-derived suppressor cells, and macrophages, among others. The review by de Visser et al[30] highlights the role of primary immune cells within the TIME and their influence on the effectiveness of tumor immunotherapy. The overall proportion, characteristics, and activation status of Th1 and Tc cells within the TIME are major factors in assessing recurrence and/or metastasis risk in CRC. B cells exert antitumor effects through antigen presentation to T cells, antibody production, and cytokine secretion. Additionally, these proteins promote tumor progression by releasing factors that cause inflammation, immunosuppression, angiogenesis, and complement activation. These dual roles make B cells both protective and oncogenic within the cancer milieu[31,32]. Tumor-associated macrophages also exert both pro- and antitumorigenic effects in the TIME, where they secrete vascular endothelial growth factor and facilitate new blood vessel formation[33,34]. Vessels and the extracellular matrix are also nonnegligible components of the TIME. Vessels are vital for nourishing tumors and maintaining tumor perfusion. They also serve as conduits for immune cells, facilitating their movement from the original tumor location to other areas, where they can contribute to immunological responses. The extracellular matrix consists of proteins, including collagen, fibronectin, elastin, and laminin. The specific composition and organization of this matrix, where tumor cells reside, can influence how immune cells interact with tumors. In certain cases, the matrix may form a physical barrier that impairs the effectiveness of antitumor immune cells.

In the recent issue of the World Journal of Clinical Oncology, Zhu et al[35] published an interesting paper titled “Identification and validation of a pyroptosis-related prognostic model for CRC based on bulk and single-cell RNA sequencing data”. The authors developed a pyroptosis-related prognostic model for CRC, confirming its correlation with immune infiltration. Specifically, the researchers accessed CRC patient data from The Cancer Genome Atlas (TCGA) database and the single-cell RNA sequencing dataset GSE178341 from the Gene Expression Omnibus to extract genes linked to pyroptosis. They further performed graphene oxide enrichment analysis on the set of pyroptosis-related genes and analyzed their differential expression. A total of 178 key genes derived from the scRNA-seq and TCGA datasets were obtained, and 8 differently expressed genes related to prognosis, CHMP2B, SDHB, BST2, UBE2D2, GJA1, AIM2, PDCD6IP, and SEZ6L2, were eventually identified. These genes were used to establish the prognostic model utilizing univariate Cox analysis and the LASSO Cox regression algorithm, and its efficacy was verified by receiver operating characteristic analysis. Additionally, the differences in immune-related functions between the high- and low-risk groups were compared using the CIBERSORTx algorithm to investigate the relationship between immune infiltration and CRC. Overall, this study may hold promise for enhancing clinical management and immunotherapy strategies for CRC patients.

In recent years, the use of three-dimensional organoid models for comprehensive evaluation of the TIME has attracted increasing amounts of attention. Neal et al[28] described an organoid methodology that may inspire subsequent research and drug discovery for studies by Zhu et al[35]. Specifically, they successfully generated patient-derived organoids (PDOs) from 100 human biopsies of surgically resected primary and metastatic tumor tissues, including the intestine, stomach, and pancreas, by plating mechanically dissociated tissue fragments in type I collagen matrix air-liquid interface (ALI) culture. They utilized WENR base medium to expand and serially passage mechanically processed tumor fragments as ALI organoids and successfully established PDOs, including CRC. This organoid model offers two main advantages: These PDOs preserve the tumor architecture and stroma expressing SMA and vimentin, indicating faithful recapitulation of the parental tumor histology, with a 73% success rate across different tumor types after one month of culture. Additionally, PDOs preserve the TIME and diverse integrated immune elements, including T cells, B cells, NK cells, and macrophages. Simultaneous analysis of gene expression and the immune repertoire in single cells suggested that the T-cell receptor repertoire is highly conserved between tumors and their corresponding PDOs. Despite these advancements, several questions remain unanswered in the study by Neal et al[36]. The exact mechanisms underlying the roles of CHMP2B and PDCD6IP, which are newly proposed prognostic biomarkers for CRC, in pyroptosis and tumors remain to be elucidated. Moreover, translating bioinformatic insights into improvements in TIME organoid model development is crucial for validating research findings and advancing personalized immunotherapy for CRC. The current investigation confirmed the predictive significance of pyroptosis-related genes and revealed that compared with other traditional prognostic models, the authors’ pyroptosis-related risk model could provide insight into personalized immunotherapy for CRC patients.

In the past decade, remarkable progress has been made in immunotherapy for CRC, which could be a successful approach for overcoming the limitations of initial diagnoses and current treatments. Yu et al[37] recently reviewed new immune-based therapies for primary and metastatic CRC. There are three major classes of immunotherapy: Immune checkpoint inhibitors (ICIs), adoptive cell transfer therapy, and tumor vaccines. Immunotherapies aimed at treating stage IV cancers using ICIs have been shown to greatly improve patient outcomes in mismatch repair-deficient CRC patients, but the use of adoptive cell transfer therapy and tumor vaccines has not yet been widely applied. In addition, monoclonal antibodies have demonstrated efficacy in treating a wide range of tumors, largely because of their favorable safety profile and ability to precisely target tumor cells. Despite these advantages, the use of monoclonal antibodies is currently limited by their high cost. Furthermore, their exquisite specificity, while beneficial in many respects, may not be optimal for combating heterogeneous tumor cell populations, including cancer stem cells, which can contribute to tumor resistance and recurrence. Thus, it is necessary to gain more insight into the appropriate and new biomarkers that will provide accurate and reliable diagnostic and prognostic information regarding the influence of the immune system on CRC. Targeting pyroptosis may reveal the potential of developing immunotherapies that treat microsatellite-stable CRC, which accounts for the majority of all CRC cases.

CLINICAL IMPLICATIONS

CRC and liver metastases from CRC are important clinically relevant issues for at least three reasons. First, CRC is the major cause of death in both men and women worldwide and in both developed and undeveloped countries. It is diagnosed more frequently due to the progressive aging of the population, obesity, and sedentarism. Second, the majority of patients die from metastatic liver disease, and only a few of the patients with colorectal liver metastases are candidates for liver resection. Third, immunotherapy, chemotherapy, and radiotherapy can potentially increase the survival time of patients with unresectable colorectal liver metastases. Extensive results have shown that CRC responds poorly to ICIs, partly because of the immunosuppressive tumor microenvironment in immunologically cold tumors, and widespread chemoresistance has led to great concern regarding their efficacy, safety, and tolerability. Studies on pyroptosis in immunotherapy have the potential to transform the way that CRC and other metastatic tumors are treated and indicate more diverse treatment approaches for CRC.

CONCLUSION

In recent years, research efforts to identify biomarkers and develop prognostic models for CRC have intensified due to their potential for improving outcome prediction. Although various strategies are being pursued, there is currently no active immunotherapy available for treating metastatic or recurrent CRC, underscoring the urgent need for more effective prognostic and predictive biomarkers. The recommendations for these patients are as follows: (1) Sufficient physical activity and weight reduction can improve cancer outcomes[38]; (2) Chemotherapy and surgical resection for patients with limited metastatic disease can play an important, and sometimes curative, role in the treatment of select patients with mCRC[39,40]; and (3) The use of 5-fluorouracil can improve survival; alternatively, aspirin can be used (indicated by a large number of epidemiological and experimental studies to be effective in the treatment of CRC). Clinicians should also recognize that combination treatment regimens of standard drugs with newer agents have been shown to improve overall survival, disease-free survival, time to progression, and quality of life compared to standard drugs alone in patients with advanced CRC. For instance, oxaliplatin plus intravenous bolus fluorouracil and leucovorin has been shown to be superior to intravenous bolus fluorouracil and leucovorin in terms of disease-free survival[41].

ACKNOWLEDGEMENTS

This study benefited from the cooperation of each researcher, whose contributions are sincerely appreciated.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Oncology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade C

Novelty: Grade C

Creativity or Innovation: Grade C

Scientific Significance: Grade C

P-Reviewer: Mathew S S-Editor: Qu XL L-Editor: Wang TQ P-Editor: Zhao YQ

References
1.  Yu P, Zhang X, Liu N, Tang L, Peng C, Chen X. Pyroptosis: mechanisms and diseases. Signal Transduct Target Ther. 2021;6:128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 996]  [Cited by in F6Publishing: 887]  [Article Influence: 295.7]  [Reference Citation Analysis (0)]
2.  Bertheloot D, Latz E, Franklin BS. Necroptosis, pyroptosis and apoptosis: an intricate game of cell death. Cell Mol Immunol. 2021;18:1106-1121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 525]  [Cited by in F6Publishing: 776]  [Article Influence: 258.7]  [Reference Citation Analysis (0)]
3.  Chan FK, Luz NF, Moriwaki K. Programmed necrosis in the cross talk of cell death and inflammation. Annu Rev Immunol. 2015;33:79-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 268]  [Article Influence: 26.8]  [Reference Citation Analysis (0)]
4.  Du T, Gao J, Li P, Wang Y, Qi Q, Liu X, Li J, Wang C, Du L. Pyroptosis, metabolism, and tumor immune microenvironment. Clin Transl Med. 2021;11:e492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 117]  [Article Influence: 39.0]  [Reference Citation Analysis (0)]
5.  Nagata S. Apoptosis and Clearance of Apoptotic Cells. Annu Rev Immunol. 2018;36:489-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 421]  [Cited by in F6Publishing: 604]  [Article Influence: 100.7]  [Reference Citation Analysis (0)]
6.  Kiraz Y, Adan A, Kartal Yandim M, Baran Y. Major apoptotic mechanisms and genes involved in apoptosis. Tumour Biol. 2016;37:8471-8486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 362]  [Article Influence: 45.3]  [Reference Citation Analysis (0)]
7.  Carneiro BA, El-Deiry WS. Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol. 2020;17:395-417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 911]  [Cited by in F6Publishing: 1191]  [Article Influence: 297.8]  [Reference Citation Analysis (0)]
8.  Majtnerová P, Roušar T. An overview of apoptosis assays detecting DNA fragmentation. Mol Biol Rep. 2018;45:1469-1478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 97]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
9.  Battistelli M, Falcieri E. Apoptotic Bodies: Particular Extracellular Vesicles Involved in Intercellular Communication. Biology (Basel). 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 220]  [Article Influence: 55.0]  [Reference Citation Analysis (0)]
10.  Wang Y, Kanneganti TD. From pyroptosis, apoptosis and necroptosis to PANoptosis: A mechanistic compendium of programmed cell death pathways. Comput Struct Biotechnol J. 2021;19:4641-4657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 199]  [Article Influence: 66.3]  [Reference Citation Analysis (0)]
11.  D'Souza CA, Heitman J. Dismantling the Cryptococcus coat. Trends Microbiol. 2001;9:112-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 45]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
12.  Broz P, Pelegrín P, Shao F. The gasdermins, a protein family executing cell death and inflammation. Nat Rev Immunol. 2020;20:143-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 449]  [Cited by in F6Publishing: 821]  [Article Influence: 164.2]  [Reference Citation Analysis (0)]
13.  Burdette BE, Esparza AN, Zhu H, Wang S. Gasdermin D in pyroptosis. Acta Pharm Sin B. 2021;11:2768-2782.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 274]  [Article Influence: 91.3]  [Reference Citation Analysis (0)]
14.  Liu X, Lieberman J. Knocking 'em Dead: Pore-Forming Proteins in Immune Defense. Annu Rev Immunol. 2020;38:455-485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 61]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
15.  Kovacs SB, Miao EA. Gasdermins: Effectors of Pyroptosis. Trends Cell Biol. 2017;27:673-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 706]  [Cited by in F6Publishing: 789]  [Article Influence: 112.7]  [Reference Citation Analysis (0)]
16.  Tsuchiya K. Switching from Apoptosis to Pyroptosis: Gasdermin-Elicited Inflammation and Antitumor Immunity. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 147]  [Article Influence: 49.0]  [Reference Citation Analysis (0)]
17.  Tsuchiya K, Nakajima S, Hosojima S, Thi Nguyen D, Hattori T, Manh Le T, Hori O, Mahib MR, Yamaguchi Y, Miura M, Kinoshita T, Kushiyama H, Sakurai M, Shiroishi T, Suda T. Caspase-1 initiates apoptosis in the absence of gasdermin D. Nat Commun. 2019;10:2091.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 287]  [Article Influence: 57.4]  [Reference Citation Analysis (0)]
18.  Matikainen S, Nyman TA, Cypryk W. Function and Regulation of Noncanonical Caspase-4/5/11 Inflammasome. J Immunol. 2020;204:3063-3069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 48]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
19.  Zhou Z, He H, Wang K, Shi X, Wang Y, Su Y, Wang Y, Li D, Liu W, Zhang Y, Shen L, Han W, Shen L, Ding J, Shao F. Granzyme A from cytotoxic lymphocytes cleaves GSDMB to trigger pyroptosis in target cells. Science. 2020;368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 344]  [Cited by in F6Publishing: 685]  [Article Influence: 171.3]  [Reference Citation Analysis (0)]
20.  Kesavardhana S, Malireddi RKS, Kanneganti TD. Caspases in Cell Death, Inflammation, and Pyroptosis. Annu Rev Immunol. 2020;38:567-595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 455]  [Article Influence: 113.8]  [Reference Citation Analysis (0)]
21.  Xue Y, Enosi Tuipulotu D, Tan WH, Kay C, Man SM. Emerging Activators and Regulators of Inflammasomes and Pyroptosis. Trends Immunol. 2019;40:1035-1052.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 169]  [Article Influence: 33.8]  [Reference Citation Analysis (0)]
22.  Wang YY, Liu XL, Zhao R. Induction of Pyroptosis and Its Implications in Cancer Management. Front Oncol. 2019;9:971.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 144]  [Article Influence: 28.8]  [Reference Citation Analysis (0)]
23.  Xia X, Wang X, Cheng Z, Qin W, Lei L, Jiang J, Hu J. The role of pyroptosis in cancer: pro-cancer or pro-"host"? Cell Death Dis. 2019;10:650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 523]  [Article Influence: 104.6]  [Reference Citation Analysis (0)]
24.  Wei X, Xie F, Zhou X, Wu Y, Yan H, Liu T, Huang J, Wang F, Zhou F, Zhang L. Role of pyroptosis in inflammation and cancer. Cell Mol Immunol. 2022;19:971-992.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 199]  [Reference Citation Analysis (0)]
25.  Fang Y, Tian S, Pan Y, Li W, Wang Q, Tang Y, Yu T, Wu X, Shi Y, Ma P, Shu Y. Pyroptosis: A new frontier in cancer. Biomed Pharmacother. 2020;121:109595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 275]  [Cited by in F6Publishing: 551]  [Article Influence: 110.2]  [Reference Citation Analysis (0)]
26.  Li L, Jiang M, Qi L, Wu Y, Song D, Gan J, Li Y, Bai Y. Pyroptosis, a new bridge to tumor immunity. Cancer Sci. 2021;112:3979-3994.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 111]  [Article Influence: 37.0]  [Reference Citation Analysis (0)]
27.  Dihlmann S, Tao S, Echterdiek F, Herpel E, Jansen L, Chang-Claude J, Brenner H, Hoffmeister M, Kloor M. Lack of Absent in Melanoma 2 (AIM2) expression in tumor cells is closely associated with poor survival in colorectal cancer patients. Int J Cancer. 2014;135:2387-2396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 86]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
28.  Miguchi M, Hinoi T, Shimomura M, Adachi T, Saito Y, Niitsu H, Kochi M, Sada H, Sotomaru Y, Ikenoue T, Shigeyasu K, Tanakaya K, Kitadai Y, Sentani K, Oue N, Yasui W, Ohdan H. Gasdermin C Is Upregulated by Inactivation of Transforming Growth Factor β Receptor Type II in the Presence of Mutated Apc, Promoting Colorectal Cancer Proliferation. PLoS One. 2016;11:e0166422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 132]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
29.  Hou J, Hsu JM, Hung MC. Molecular mechanisms and functions of pyroptosis in inflammation and antitumor immunity. Mol Cell. 2021;81:4579-4590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 133]  [Article Influence: 44.3]  [Reference Citation Analysis (0)]
30.  de Visser KE, Joyce JA. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell. 2023;41:374-403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 502]  [Reference Citation Analysis (0)]
31.  Yuen GJ, Demissie E, Pillai S. B lymphocytes and cancer: a love-hate relationship. Trends Cancer. 2016;2:747-757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 258]  [Article Influence: 43.0]  [Reference Citation Analysis (0)]
32.  Laumont CM, Banville AC, Gilardi M, Hollern DP, Nelson BH. Tumour-infiltrating B cells: immunological mechanisms, clinical impact and therapeutic opportunities. Nat Rev Cancer. 2022;22:414-430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 140]  [Article Influence: 70.0]  [Reference Citation Analysis (0)]
33.  Güç E, Pollard JW. Redefining macrophage and neutrophil biology in the metastatic cascade. Immunity. 2021;54:885-902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 64]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
34.  DeNardo DG, Ruffell B. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol. 2019;19:369-382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1310]  [Cited by in F6Publishing: 1301]  [Article Influence: 260.2]  [Reference Citation Analysis (0)]
35.  Zhu LH, Yang J, Zhang YF, Yan L, Lin WR, Liu WQ. Identification and validation of a pyroptosis-related prognostic model for colorectal cancer based on bulk and single-cell RNA sequencing data. World J Clin Oncol. 2024;15:329-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (1)]
36.  Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, Liu IH, Chiou SH, Salahudeen AA, Smith AR, Deutsch BC, Liao L, Zemek AJ, Zhao F, Karlsson K, Schultz LM, Metzner TJ, Nadauld LD, Tseng YY, Alkhairy S, Oh C, Keskula P, Mendoza-Villanueva D, De La Vega FM, Kunz PL, Liao JC, Leppert JT, Sunwoo JB, Sabatti C, Boehm JS, Hahn WC, Zheng GXY, Davis MM, Kuo CJ. Organoid Modeling of the Tumor Immune Microenvironment. Cell. 2018;175:1972-1988.e16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 792]  [Cited by in F6Publishing: 811]  [Article Influence: 135.2]  [Reference Citation Analysis (0)]
37.  Yu I, Dakwar A, Takabe K. Immunotherapy: Recent Advances and Its Future as a Neoadjuvant, Adjuvant, and Primary Treatment in Colorectal Cancer. Cells. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
38.  Davies NJ, Batehup L, Thomas R. The role of diet and physical activity in breast, colorectal, and prostate cancer survivorship: a review of the literature. Br J Cancer. 2011;105 Suppl 1:S52-S73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 178]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
39.  Abdalla EK, Vauthey JN, Ellis LM, Ellis V, Pollock R, Broglio KR, Hess K, Curley SA. Recurrence and outcomes following hepatic resection, radiofrequency ablation, and combined resection/ablation for colorectal liver metastases. Ann Surg. 2004;239:818-25; discussion 825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1364]  [Cited by in F6Publishing: 1251]  [Article Influence: 62.6]  [Reference Citation Analysis (0)]
40.  Charnsangavej C, Clary B, Fong Y, Grothey A, Pawlik TM, Choti MA. Selection of patients for resection of hepatic colorectal metastases: expert consensus statement. Ann Surg Oncol. 2006;13:1261-1268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 227]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
41.  Akhtar R, Chandel S, Sarotra P, Medhi B. Current status of pharmacological treatment of colorectal cancer. World J Gastrointest Oncol. 2014;6:177-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (1)]