BPG is committed to discovery and dissemination of knowledge
Editorial Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Nov 26, 2025; 13(33): 113216
Published online Nov 26, 2025. doi: 10.12998/wjcc.v13.i33.113216
When new therapies arrive, we should think more: The unseen challenges of immunotherapy-induced remodeling
Rui Gang Wang, Department of Gastroenterology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing 102218, China
ORCID number: Rui-Gang Wang (0000-0002-9053-5329).
Author contributions: Wang RG contributed to the design, drafting, revision, and review of the manuscript content.
Supported by Beijing Tsinghua Changgung Hospital Youth Fund, No. 12021C1011.
Conflict-of-interest statement: The author reports no relevant conflicts of interest for this article.
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: Rui Gang Wang, Department of Gastroenterology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, No. 168 Litang Road, Changping District, Beijing 102218, China. doctorwrg@163.com
Received: August 19, 2025
Revised: August 30, 2025
Accepted: October 27, 2025
Published online: November 26, 2025
Processing time: 94 Days and 13.3 Hours

Abstract

The paradigm-shifting efficacy of immune checkpoint inhibitors in microsatellite instability-high colorectal cancer demands a critical appraisal of their long-term tissue effects. This editorial is a landmark case report revealing spontaneous colonic transection following pathological complete response to pembrolizumab - a first-in-medicine finding. Despite radiological resolution of disease, profound immune-mediated tissue remodeling resulted in catastrophic structural failure at the tumor site. This phenomenon exposes critical limitations in current imaging modalities to detect immunotherapy-induced bowel wall fragility and necessitates heightened awareness among surgeons. As immune checkpoint inhibitors move into neoadjuvant settings with rising complete response rates, we must reassess surgical planning, consider prophylactic interventions for high-risk anatomy, and develop biomarkers for tissue integrity. This case underscores that tumor regression does not equate to restored organ function, urging multidisciplinary vigilance against delayed structural toxicity.

Key Words: Immune checkpoint inhibitors; Microsatellite instability-high colorectal cancer; Tissue remodeling; Pathological complete response; Toxicity

Core Tip: The unprecedented case of spontaneous colonic transection after pathological complete response to pembrolizumab in microsatellite instability-high colorectal cancer highlights a critical blind spot in immune oncology. While immune checkpoint inhibitors achieve remarkable tumor regression, they can induce profound bowel wall damage invisible to conventional imaging. This editorial calls for urgent multidisciplinary action: Re-evaluating surgical timing and techniques in immunotherapy responders, developing novel biomarkers to predict bowel wall integrity, and implementing enhanced surveillance protocols to prevent catastrophic complications from delayed tissue toxicity.



INTRODUCTION

Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that induce or amplify anti-tumor T-cell responses by blocking inhibitory immune-regulatory pathways such as programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and their ligands (PD-L1). By antagonizing ligand-receptor pairs that physiologically restrain T-cell activation, ICIs convert immunologically “cold” tumors into “hot” ones, thereby enabling durable immune-mediated tumor control. The advent of ICIs that target PD-1/PD-L1 and CTLA-4 has irrevocably altered the therapeutic landscape for microsatellite instability-high (MSI-H) or deficient mismatch repair colorectal cancers (CRC). Groundbreaking trials have established pembrolizumab and nivolumab (both of which are anti-PD-1 antibodies) and ipilimumab (an anti-CTLA-4 antibody) - either alone or in combination - as superior to traditional chemotherapy for metastatic CRC, with significantly improved progression-free and overall survival and more favorable toxicity profiles[1-3]. This success has propelled ICIs into earlier disease settings (including as a neoadjuvant therapy for locally advanced tumors) where they have demonstrated remarkable pathological complete response (pCR) rates exceeding 60% in some studies[4-6]. A pCR is defined as the absence of any viable tumor cells in surgical specimens following treatment (ypT0N0). The allure of non-operative management or organ-sparing surgery following a deep clinical response is understandably strong[7,8]. However, an extraordinary case reported by Lee et al[9] (which details a spontaneous colonic transection during surgery after pCR to pembrolizumab) sounds a crucial alarm. This unprecedented complication forces us to confront the sobering reality that the immune system’s ability to eradicate cancer can simultaneously inflict profound, potentially catastrophic damage on normal tissue architecture. Such effects may remain hidden until surgical intervention or a catastrophic clinical crisis ensues.

THE CASE: A PARADIGM-SHIFTING COMPLICATION

Lee et al[9] presented a meticulously documented case of a 44-year-old male with bulky, locally advanced MSI-H transverse colon cancer and synchronous liver metastases. Following a diverting ileostomy, he received 36 cycles of pembrolizumab over approximately two years, showing a sustained clinical and radiological complete response. A subsequent laparoscopic right hemicolectomy (intended as a curative resection) revealed a startling finding, namely, a complete spontaneous transection of the colon at the original tumor site. Histopathology confirmed a pCR with extensive transmural fibrosis, calcification, microvascular damage, and chronic inflammation, but no residual tumor. This structural discontinuity occurred despite a lack of evidence of any impending perforation or obstruction on recent radiological scans. This represents the first documented case of complete bowel wall discontinuity following an ICI-induced pCR beyond any previously reported obstructions or fibrotic strictures[10].

BEYOND TUMOR KILLING: THE DOUBLE-EDGED SWORD OF IMMUNE-MEDIATED TISSUE REMODELING

The pathological findings in this case point toward intense and sustained immune activity within the bowel wall. ICIs work by activating latent anti-tumor T-cell responses. However, this activation is not tumor-specific. T-cell activation and its associated inflammatory cascade (which involves cytokines such as interferon-gamma, tumor necrosis factor alpha, and interleukin-17) can indiscriminately target stromal and endothelial cells within the tumor microenvironment and adjacent normal tissue[11,12]. While acute immune-related adverse events (irAEs) such as colitis are highly recognizable and often manageable via immunosuppression, the case reported by Lee et al[9] reveals a more insidious, long-term consequence: Chronic, immune-driven tissue remodeling.

This remodeling involves complex biological processes (Figure 1), including: (1) Fibrosis: Persistent inflammation triggers fibroblast activation and excessive extracellular matrix (ECM) deposition[13]. Although this is initially a reparative response, dysregulated fibrosis replaces functional tissue with stiff, acellular collagen, which compromises tensile strength and elasticity. Transforming growth factor-beta is a key driver of this process[14]; (2) Microvascular damage: Immune-mediated endothelial injury disrupts the microvasculature, resulting in ischemia and hypoxia[15]. These further fuel inflammation and fibrosis in a vicious cycle. Angiogenesis is often impaired in these scenarios[16]; (3) Altered matrix metalloproteinase (MMP) activity: Immune cells secrete MMPs that degrade ECM components. An imbalance between MMPs and their inhibitors can lead to excessive matrix breakdown, thereby weakening the tissue scaffold[17]; and (4) Impaired healing: In combination with these processes, chronic inflammation continually disrupts normal tissue repair mechanisms, resulting in the formation of structurally unsound scar tissue[18].

Figure 1
Figure 1 Pathogenesis of immune-mediated bowel wall remodeling. Stage 1 (blue): Immune checkpoint inhibition is lifted, activating and infiltrating T cells; stage 2 (orange): Activated T cells and M1-type macrophages release large amounts of interferon-gamma and tumor necrosis factor alpha; stage 3 (green): Fibroblasts are activated and upregulate transforming growth factor beta, initiating the fibrosis program; stage 4 (red): Transforming growth factor beta promotes the deposition of type I/III collagen; simultaneously, inflammatory mediators cause vascular endothelial damage, microthrombosis formation, tissue hypoxia, and impaired angiogenesis; stage 5 (purple): Disordered collagen deposition disrupts the matrix structure, leading to reduced tissue tension and ultimately increased intestinal wall fragility. PD: Programmed death; IFN-γ: Interferon-gamma; TNF-α: Tumor necrosis factor alpha; TGF-β: Transforming growth factor beta.

These processes can architecturally compromise the bowel wall, rendering it fragile and failure-prone (despite it being tumor-free). This condition cannot be adequately detected using current standard imaging modalities, such as computed tomography (CT), magnetic resonance imaging, or even positron emission tomography-CT. These imaging techniques primarily assess tumor burden and gross morphology rather than the microscopic integrity or mechanical strength of the organ tissue[19]. It is critical to acknowledge the potential contributory role of prior chemotherapy, a common sequence in many cancer treatments. Although this particular patient received pembrolizumab as a primary treatment, many patients with MSI-H CRC receive chemotherapy before immunotherapy is considered. Cytotoxic chemotherapy agents are known to cause direct damage to the gastrointestinal mucosa and alter the tumor microenvironment by inducing stromal injury, promoting a pro-fibrotic state, and increasing epithelial permeability[20]. This chemotherapy-induced “priming” of the tissue could potentially lower the threshold for or exacerbate subsequent immune-mediated injury upon ICI administration, leading to more severe remodeling and fragility. The interaction between chemotherapy-induced damage and immunotherapy-driven inflammation warrants further mechanistic investigation.

SURGICAL PLANNING IN THE ICI ERA: A CALL FOR REASSESSMENT

The United States Food and Drug Administration approved ipilimumab in 2011 as the first CTLA-4 inhibitor for the treatment of metastatic melanoma. The ICI era is not merely a temporal label, but a systemic transformation in cancer treatment. It marks a shift from purely cytotoxic paradigms to immune-centric treatment sequencing, introduces novel response patterns that challenge conventional radiographic and pathologic endpoints, and necessitates the recalibration of surgical timing based on immune-related toxicities and more favorable survivability. This landmark case necessitates an urgent reevaluation of surgical strategies for patients exhibiting major responses to neoadjuvant or definitive ICI therapies, particularly in MSI-H CRC (Figure 2).

Figure 2
Figure 2 Surgical risk stratification algorithm. Evidence-based decision pathway for resection planning in immune checkpoint inhibitor-treated patients. High-risk criteria (red) include: > 12 immune checkpoint inhibitor cycles, initial tumor > 5 cm, or radiologic fibrosis signs (loss of mural stratification, SUVmax 2-4 on fluorodeoxyglucose-positron emission tomography, or elastography stiffness > 8 kPa). Mandatory intraoperative indocyanine green perfusion assessment guides anastomotic safety: Segments with perfusion delay > 30 seconds or bursting pressure < 25 mmHg require diversion. Prophylactic stoma is indicated when ≥ 2 major risk factors coexist. ICI: Immune checkpoint inhibitor; ICG: Indocyanine green.

The reassessment of surgical planning should include: (1) Heightened preoperative suspicion: Surgeons must be acutely aware that radiological evidence of clinical complete response and the disappearance of all visible tumors don’t necessarily equate to a structurally normal bowel wall and may not be predictive of pCR. A history of prolonged ICI therapy (such as the 36 cycles reported in this case), prior irAEs (especially colitis), or bulky initial tumors causing significant desmoplasia should raise concern about potential bowel wall fragility[10,21]; (2) Reconsidering surgical timing and the necessity of resection: Although organ preservation is desirable, this case highlights that leaving a heavily pretreated, potentially fibrotic bowel segment in situ is associated with an undetermined risk of delayed perforation or stricture. Surgery itself also poses risks to fragile tissue. The optimal timing of surgery post-ICI cessation remains unknown and requires thorough investigation. However, resection should typically be performed earlier in responders to mitigate the risks associated with prolonged tissue remodeling[22]; (3) Intraoperative vigilance and technique: Laparoscopic entry and dissection near the tumor site warrant extreme caution, and gentle tissue handling is paramount. Surgeons should meticulously inspect the bowel segment that harbored the initial tumor for any signs of thinning, fibrosis, or discontinuity before surgical mobilization. A wider margin of apparently normal bowel tissue proximal and distal to the fibrotic segment should be considered for resection to ensure anastomotic security. Techniques such as indocyanine green perfusion assessment may be valuable in this assessment[23]; (4) Prophylaxis: In high-risk scenarios (e.g., evidence of extensive fibrosis on preoperative imaging, long ICI treatment duration, prior obstruction/irAE, or intraoperative signs of significant bowel wall fragility), prophylactic stoma formation should be strongly considered even when anastomosis appears technically feasible. This provides a critical safety net should anastomotic failure occur due to poor tissue quality. Temporary diversion may be preferable to catastrophic leaks[24]; and (5) Pathology: Close communication with pathologists is essential. Beyond tumor staging, a detailed histological assessment of the resected bowel wall is crucial to characterize the extent of fibrosis, inflammation, and vascular changes. Special stains (e.g., trichrome for collagen, CD31/CD34 for vasculature) and molecular analysis (e.g., immune cell profiling, fibrosis markers) can provide insight into the mechanisms of the tissue damage[25].

MORE THAN THE SCALPEL: MULTIDISCIPLINARY IMPERATIVES AND FUTURE DIRECTIONS

Addressing this emerging challenge extends well beyond the operating room and demands a concerted multidisciplinary effort (Figure 3). First, there is an urgent need for biomarkers to identify patients at high risk for severe tissue remodeling to avoid catastrophic events. Potential biomarker candidates include: (1) Circulating markers: Serum/plasma levels of ECM turnover products (e.g., N-terminal type III collagen, C-terminal type VIa3 collagen), inflammatory cytokines (e.g., interleukin-6, tumor necrosis factor alpha), or markers of endothelial damage (e.g., vascular cellular adhesion molecule-1, angiopoietin-2)[26,27]; (2) Imaging biomarkers: Advanced magnetic resonance imaging techniques (e.g., magnetization transfer imaging, diffusion kurtosis imaging) or positron emission tomography tracers that target fibrosis (e.g., 68Ga-FAPI-04) or inflammation could be used to detect microstructural changes that precede catastrophic failure[28,29]; and (3) Tissue-based biomarkers: Analyzing pretreatment biopsies for specific immune cell infiltrates (e.g., high CD8+/Treg ratios, macrophage polarization) or stromal signatures could predict the severity of subsequent tissue remodeling[30].

Figure 3
Figure 3 Multidisciplinary risk mitigation framework. Translational framework for predicting and mitigating bowel wall fragility across treatment phases. Pre-treatment: Stromal gene signatures (Type III collagen/Matrix metallopeptidase 7 ratio) from biopsies predict remodeling propensity. On-treatment: Serum N-terminal type III collagen (type III collagen fragment) > 25 ng/mL and 68Ga-FAPI positron emission tomography SUVmax > 6 signal active fibrosis. Surgical: Assess the biological resilience of intestinal wall tissue. Post-op: AI-powered digital pathology (collagen orientation index) informs stoma reversal timing. MMR: Mismatch repair; MSI: Microsatellite instability; PRO-C3: N-terminal propeptide of collagen type 3; IL-6: Interleukin-6; PET: Positron emission tomography; ICG: Indocyanine green; AI: Artificial intelligence.

Second, imaging surveillance strategies need to be refined. The standard response evaluation criteria for solid tumors are insufficient. Radiologists require specific training and protocols for scrutinizing the bowel wall at the initial tumor site for subtle signs of wall thickening, fat stranding, loss of mural stratification, or pneumatosis - even in the absence of residual mass - that might indicate fragility[19].

Third, mitigating strategies should be explored thoroughly. Research is needed to confirm whether concurrent or sequential therapies can modulate fibrosis without compromising anti-tumor efficacy. Potential approaches include short courses of antifibrotics (e.g., pirfenidone, nintedanib) after a tumor response is achieved, or the use of agents that target specific fibrotic pathways (e.g., transforming growth factor-beta inhibitors)[31]. The role of microbiome modulation in preventing chronic inflammation also warrants investigation[32].

Fourth, the roles of alternative immune checkpoints such as lymphocyte-activation gene 3, T-cell immunoglobulin and mucin-domain containing-3, and T-cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibition motif domains must be evaluated[33]. Primary and acquired resistance to PD-1/CTLA-4 blockade is often mediated by the compensatory upregulation of these alternative immune checkpoints. Novel agents that target these pathways are currently entering clinical practice to address immune checkpoint blockade resistance and enhance anti-tumor immunity. However, the impact of these agents on immune-mediated tissue remodeling remains unknown. Dual checkpoint blockade could potentially amplify off-target tissue damage. Conversely, more precise targeting could potentially reduce it. Therefore, future research should concurrently evaluate the efficacy and tissue-specific toxicity profiles of these next-generation immunotherapies.

Finally, there is a need for prospective registries and trials involving a systematic collection of data on surgical complications, tissue pathology, and long-term outcomes in patients treated with ICI as they undergo surgery. Dedicated clinical trials are needed to compare different surgical approaches (e.g., resection vs watchful waiting in clinical complete response, either with or without diversion) and to evaluate potential protective pharmacologic interventions.

CONCLUSION

This remarkable case of spontaneous colonic transection following pembrolizumab-induced pCR is not merely an anomaly; rather, it is a sentinel event that reveals a previously underappreciated dimension of ICI toxicity. It shatters the assumption that tumor regression equates to restored organ integrity. Potent immune activation that drives tumor cell death can also induce parallel processes (i.e., fibrosis, vascular damage, and impaired healing) that silently undermine the mechanical strength of the bowel wall. Current imaging modalities provide an alarmingly incomplete picture of this risk. As ICIs become a standard neoadjuvant therapy for MSI-H CRC with organ preservation as an enticing goal, this case imposes a critical mandate for caution and adaptation. Surgeons must therefore approach post-ICI bowel surgery with heightened suspicion for fragility, modify their techniques accordingly, and embrace prophylactic stomas when elevated risk is established. The broader oncology community must prioritize the development of biomarkers to predict tissue vulnerability and novel imaging techniques to visualize it. Multidisciplinary collaboration is essential to achieve this goal. The benefits of immunotherapy should not be overshadowed by preventable structural complications. Recognizing and mitigating the delayed consequences of immune-mediated tissue remodeling represents the next frontier in improving safety and long-term outcomes for patients who benefit from these groundbreaking agents. Although the eradication of tumors is a triumph, the integrity of the surviving organ remains paramount.

Footnotes

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

Peer-review model: Single blind

Corresponding Author’s Membership in Professional Societies: Chinese Medical Education Association Committee for the Promotion of Basic and Clinical Research, CPBCR-0195; Member of the Digestive Endoscopy Branch of the Cross-Strait Medical and Health Exchange Association, ZXHNJ-1-163.

Specialty type: Medicine, research and experimental

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B, Grade B

Novelty: Grade B, Grade B

Creativity or Innovation: Grade B, Grade B

Scientific Significance: Grade B, Grade B

P-Reviewer: Tahri A, Post Doctoral Researcher, Italy S-Editor: Bai SR L-Editor: A P-Editor: Yu HG

References
1.  André T, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, Smith D, Garcia-Carbonero R, Benavides M, Gibbs P, de la Fouchardiere C, Rivera F, Elez E, Bendell J, Le DT, Yoshino T, Van Cutsem E, Yang P, Farooqui MZH, Marinello P, Diaz LA Jr; KEYNOTE-177 Investigators. Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. N Engl J Med. 2020;383:2207-2218.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 962]  [Cited by in RCA: 1981]  [Article Influence: 330.2]  [Reference Citation Analysis (0)]
2.  André T, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, Morse MA, Van Cutsem E, McDermott R, Hill A, Sawyer MB, Hendlisz A, Neyns B, Abdullaev S, Memaj A, Lei M, Dixon M, Kopetz S, Overman MJ. Nivolumab plus low-dose ipilimumab in previously treated patients with microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: 4-year follow-up from CheckMate 142. Ann Oncol. 2022;33:1052-1060.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 168]  [Article Influence: 42.0]  [Reference Citation Analysis (0)]
3.  Lenz HJ, Van Cutsem E, Luisa Limon M, Wong KYM, Hendlisz A, Aglietta M, García-Alfonso P, Neyns B, Luppi G, Cardin DB, Dragovich T, Shah U, Abdullaev S, Gricar J, Ledeine JM, Overman MJ, Lonardi S. First-Line Nivolumab Plus Low-Dose Ipilimumab for Microsatellite Instability-High/Mismatch Repair-Deficient Metastatic Colorectal Cancer: The Phase II CheckMate 142 Study. J Clin Oncol. 2022;40:161-170.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 60]  [Cited by in RCA: 464]  [Article Influence: 116.0]  [Reference Citation Analysis (0)]
4.  Chalabi M, Fanchi LF, Dijkstra KK, Van den Berg JG, Aalbers AG, Sikorska K, Lopez-Yurda M, Grootscholten C, Beets GL, Snaebjornsson P, Maas M, Mertz M, Veninga V, Bounova G, Broeks A, Beets-Tan RG, de Wijkerslooth TR, van Lent AU, Marsman HA, Nuijten E, Kok NF, Kuiper M, Verbeek WH, Kok M, Van Leerdam ME, Schumacher TN, Voest EE, Haanen JB. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat Med. 2020;26:566-576.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 341]  [Cited by in RCA: 978]  [Article Influence: 163.0]  [Reference Citation Analysis (0)]
5.  Ludford K, Ho WJ, Thomas JV, Raghav KPS, Murphy MB, Fleming ND, Lee MS, Smaglo BG, You YN, Tillman MM, Kamiya-Matsuoka C, Thirumurthi S, Messick C, Johnson B, Vilar E, Dasari A, Shin S, Hernandez A, Yuan X, Yang H, Foo WC, Qiao W, Maru D, Kopetz S, Overman MJ. Neoadjuvant Pembrolizumab in Localized Microsatellite Instability High/Deficient Mismatch Repair Solid Tumors. J Clin Oncol. 2023;41:2181-2190.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 75]  [Cited by in RCA: 119]  [Article Influence: 39.7]  [Reference Citation Analysis (0)]
6.  Cercek A, Lumish M, Sinopoli J, Weiss J, Shia J, Lamendola-Essel M, El Dika IH, Segal N, Shcherba M, Sugarman R, Stadler Z, Yaeger R, Smith JJ, Rousseau B, Argiles G, Patel M, Desai A, Saltz LB, Widmar M, Iyer K, Zhang J, Gianino N, Crane C, Romesser PB, Pappou EP, Paty P, Garcia-Aguilar J, Gonen M, Gollub M, Weiser MR, Schalper KA, Diaz LA Jr. PD-1 Blockade in Mismatch Repair-Deficient, Locally Advanced Rectal Cancer. N Engl J Med. 2022;386:2363-2376.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 989]  [Cited by in RCA: 1028]  [Article Influence: 257.0]  [Reference Citation Analysis (0)]
7.  Cercek A, Foote MB, Rousseau B, Smith JJ, Shia J, Sinopoli J, Weiss J, Lumish M, Temple L, Patel M, Wilde C, Saltz LB, Argiles G, Stadler Z, Artz O, Maron S, Ku G, Gu P, Janjigian YY, Molena D, Iyer G, Coleman J, Abida W, Cohen S, Soares K, Schattner M, Strong VE, Yaeger R, Paty P, Shcherba M, Sugarman R, Romesser PB, Zervoudakis A, Desai A, Segal NH, El Dika I, Widmar M, Wei I, Pappou E, Fumo G, Aparo S, Gonen M, Gollub M, Jayaprakasam VS, Kim TH, Garcia Aguilar J, Weiser M, Diaz LA Jr. Nonoperative Management of Mismatch Repair-Deficient Tumors. N Engl J Med. 2025;392:2297-2308.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 45]  [Cited by in RCA: 50]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
8.  Bando H, Tsukada Y, Inamori K, Togashi Y, Koyama S, Kotani D, Fukuoka S, Yuki S, Komatsu Y, Homma S, Taketomi A, Uemura M, Kato T, Fukui M, Wakabayashi M, Nakamura N, Kojima M, Kawachi H, Kirsch R, Yoshida T, Suzuki Y, Sato A, Nishikawa H, Ito M, Yoshino T. Preoperative Chemoradiotherapy plus Nivolumab before Surgery in Patients with Microsatellite Stable and Microsatellite Instability-High Locally Advanced Rectal Cancer. Clin Cancer Res. 2022;28:1136-1146.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 121]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
9.  Lee C, Kim MH, Choi ET, Park IJ, Lim SB, Yoon YS, Kim CW, Lee JL, Park EJ. Spontaneous colonic transection following pathologic complete response to pembrolizumab in high microsatellite instability colorectal cancer: A case report and review of literature. World J Clin Cases. 2025;13:110330.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
10.  Platt JR, Allotey J, Alouani E, Glasbey J, Intini R, Lonardi S, Mazzoli G, Militello AM, Modest DP, Palle J, Pietrantonio F, Riyad K, Samuel L, Schulze AV, Shiu KK, Taieb J, Tolan DJM, West NP, Westwood AC, Williams CJM, Seligmann JF. Risk of bowel obstruction in patients with colon cancer responding to immunotherapy: an international case series. ESMO Open. 2024;9:103698.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
11.  Fujiwara Y, Horita N, Adib E, Zhou S, Nassar AH, Asad ZUA, Cortellini A, Naqash AR. Treatment-related adverse events, including fatal toxicities, in patients with solid tumours receiving neoadjuvant and adjuvant immune checkpoint blockade: a systematic review and meta-analysis of randomised controlled trials. Lancet Oncol. 2024;25:62-75.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 39]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
12.  Postow MA, Sidlow R, Hellmann MD. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N Engl J Med. 2018;378:158-168.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2308]  [Cited by in RCA: 3379]  [Article Influence: 422.4]  [Reference Citation Analysis (0)]
13.  Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012;18:1028-1040.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2470]  [Cited by in RCA: 2743]  [Article Influence: 195.9]  [Reference Citation Analysis (0)]
14.  Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-β: the master regulator of fibrosis. Nat Rev Nephrol. 2016;12:325-338.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1512]  [Cited by in RCA: 2683]  [Article Influence: 268.3]  [Reference Citation Analysis (0)]
15.  Jain RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell. 2014;26:605-622.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1120]  [Cited by in RCA: 1232]  [Article Influence: 102.7]  [Reference Citation Analysis (0)]
16.  Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011;473:298-307.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3471]  [Cited by in RCA: 4110]  [Article Influence: 274.0]  [Reference Citation Analysis (0)]
17.  de Almeida LGN, Thode H, Eslambolchi Y, Chopra S, Young D, Gill S, Devel L, Dufour A. Matrix Metalloproteinases: From Molecular Mechanisms to Physiology, Pathophysiology, and Pharmacology. Pharmacol Rev. 2022;74:712-768.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 213]  [Cited by in RCA: 278]  [Article Influence: 69.5]  [Reference Citation Analysis (0)]
18.  Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med. 2014;6:265sr6.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2114]  [Cited by in RCA: 2272]  [Article Influence: 189.3]  [Reference Citation Analysis (0)]
19.  Sala E, Mema E, Himoto Y, Veeraraghavan H, Brenton JD, Snyder A, Weigelt B, Vargas HA. Unravelling tumour heterogeneity using next-generation imaging: radiomics, radiogenomics, and habitat imaging. Clin Radiol. 2017;72:3-10.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 218]  [Cited by in RCA: 264]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
20.  Chrysostomou D, Roberts LA, Marchesi JR, Kinross JM. Gut Microbiota Modulation of Efficacy and Toxicity of Cancer Chemotherapy and Immunotherapy. Gastroenterology. 2023;164:198-213.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 144]  [Article Influence: 48.0]  [Reference Citation Analysis (0)]
21.  André T, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt CJA, Smith D, Garcia-Carbonero R, Alcaide-Garcia J, Gibbs P, de la Fouchardiere C, Rivera F, Elez E, Le DT, Yoshino T, Zuo Y, Fogelman D, Adelberg D, Diaz LA. Pembrolizumab versus chemotherapy in microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer: 5-year follow-up from the randomized phase III KEYNOTE-177 study. Ann Oncol. 2025;36:277-284.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 37]  [Article Influence: 37.0]  [Reference Citation Analysis (0)]
22.  Taieb J, Svrcek M, Cohen R, Basile D, Tougeron D, Phelip JM. Deficient mismatch repair/microsatellite unstable colorectal cancer: Diagnosis, prognosis and treatment. Eur J Cancer. 2022;175:136-157.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 149]  [Cited by in RCA: 175]  [Article Influence: 43.8]  [Reference Citation Analysis (0)]
23.  Jafari MD, Pigazzi A, McLemore EC, Mutch MG, Haas E, Rasheid SH, Wait AD, Paquette IM, Bardakcioglu O, Safar B, Landmann RG, Varma MG, Maron DJ, Martz J, Bauer JJ, George VV, Fleshman JW Jr, Steele SR, Stamos MJ. Perfusion Assessment in Left-Sided/Low Anterior Resection (PILLAR III): A Randomized, Controlled, Parallel, Multicenter Study Assessing Perfusion Outcomes With PINPOINT Near-Infrared Fluorescence Imaging in Low Anterior Resection. Dis Colon Rectum. 2021;64:995-1002.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 39]  [Cited by in RCA: 113]  [Article Influence: 22.6]  [Reference Citation Analysis (1)]
24.  Benson AB, Venook AP, Adam M, Chang G, Chen YJ, Ciombor KK, Cohen SA, Cooper HS, Deming D, Garrido-Laguna I, Grem JL, Haste P, Hecht JR, Hoffe S, Hunt S, Hussan H, Johung KL, Joseph N, Kirilcuk N, Krishnamurthi S, Malla M, Maratt JK, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Nurkin S, Overman MJ, Parikh A, Patel H, Pedersen K, Saltz L, Schneider C, Shibata D, Shogan B, Skibber JM, Sofocleous CT, Tavakkoli A, Willett CG, Wu C, Gurski LA, Snedeker J, Jones F. Colon Cancer, Version 3.2024, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2024;22:e240029.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 37]  [Cited by in RCA: 114]  [Article Influence: 57.0]  [Reference Citation Analysis (0)]
25.  Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov. 2019;18:197-218.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1151]  [Cited by in RCA: 2349]  [Article Influence: 335.6]  [Reference Citation Analysis (0)]
26.  Leeming DJ, Karsdal MA, Byrjalsen I, Bendtsen F, Trebicka J, Nielsen MJ, Christiansen C, Møller S, Krag A. Novel serological neo-epitope markers of extracellular matrix proteins for the detection of portal hypertension. Aliment Pharmacol Ther. 2013;38:1086-1096.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 73]  [Cited by in RCA: 79]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
27.  Henderson NC, Rieder F, Wynn TA. Fibrosis: from mechanisms to medicines. Nature. 2020;587:555-566.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 266]  [Cited by in RCA: 1251]  [Article Influence: 208.5]  [Reference Citation Analysis (0)]
28.  Friedrich-Rust M, Poynard T, Castera L. Critical comparison of elastography methods to assess chronic liver disease. Nat Rev Gastroenterol Hepatol. 2016;13:402-411.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 160]  [Cited by in RCA: 191]  [Article Influence: 19.1]  [Reference Citation Analysis (0)]
29.  Hathi DK, Jones EF. (68)Ga FAPI PET/CT: Tracer Uptake in 28 Different Kinds of Cancer. Radiol Imaging Cancer. 2019;1:e194003.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 50]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
30.  Pagès F, Mlecnik B, Marliot F, Bindea G, Ou FS, Bifulco C, Lugli A, Zlobec I, Rau TT, Berger MD, Nagtegaal ID, Vink-Börger E, Hartmann A, Geppert C, Kolwelter J, Merkel S, Grützmann R, Van den Eynde M, Jouret-Mourin A, Kartheuser A, Léonard D, Remue C, Wang JY, Bavi P, Roehrl MHA, Ohashi PS, Nguyen LT, Han S, MacGregor HL, Hafezi-Bakhtiari S, Wouters BG, Masucci GV, Andersson EK, Zavadova E, Vocka M, Spacek J, Petruzelka L, Konopasek B, Dundr P, Skalova H, Nemejcova K, Botti G, Tatangelo F, Delrio P, Ciliberto G, Maio M, Laghi L, Grizzi F, Fredriksen T, Buttard B, Angelova M, Vasaturo A, Maby P, Church SE, Angell HK, Lafontaine L, Bruni D, El Sissy C, Haicheur N, Kirilovsky A, Berger A, Lagorce C, Meyers JP, Paustian C, Feng Z, Ballesteros-Merino C, Dijkstra J, van de Water C, van Lent-van Vliet S, Knijn N, Mușină AM, Scripcariu DV, Popivanova B, Xu M, Fujita T, Hazama S, Suzuki N, Nagano H, Okuno K, Torigoe T, Sato N, Furuhata T, Takemasa I, Itoh K, Patel PS, Vora HH, Shah B, Patel JB, Rajvik KN, Pandya SJ, Shukla SN, Wang Y, Zhang G, Kawakami Y, Marincola FM, Ascierto PA, Sargent DJ, Fox BA, Galon J. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet. 2018;391:2128-2139.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1567]  [Cited by in RCA: 1540]  [Article Influence: 192.5]  [Reference Citation Analysis (0)]
31.  Distler JHW, Györfi AH, Ramanujam M, Whitfield ML, Königshoff M, Lafyatis R. Shared and distinct mechanisms of fibrosis. Nat Rev Rheumatol. 2019;15:705-730.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 187]  [Cited by in RCA: 463]  [Article Influence: 66.1]  [Reference Citation Analysis (0)]
32.  Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, Fidelle M, Flament C, Poirier-Colame V, Opolon P, Klein C, Iribarren K, Mondragón L, Jacquelot N, Qu B, Ferrere G, Clémenson C, Mezquita L, Masip JR, Naltet C, Brosseau S, Kaderbhai C, Richard C, Rizvi H, Levenez F, Galleron N, Quinquis B, Pons N, Ryffel B, Minard-Colin V, Gonin P, Soria JC, Deutsch E, Loriot Y, Ghiringhelli F, Zalcman G, Goldwasser F, Escudier B, Hellmann MD, Eggermont A, Raoult D, Albiges L, Kroemer G, Zitvogel L. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91-97.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2493]  [Cited by in RCA: 4019]  [Article Influence: 446.6]  [Reference Citation Analysis (0)]
33.  Cai L, Li Y, Tan J, Xu L, Li Y. Targeting LAG-3, TIM-3, and TIGIT for cancer immunotherapy. J Hematol Oncol. 2023;16:101.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 55]  [Cited by in RCA: 208]  [Article Influence: 69.3]  [Reference Citation Analysis (0)]