Treatment of immune checkpoint inhibitor-related colitis: a narrative review
Introduction
Immunotherapy refers to a broad range of methods for treating cancer that generate or boost the immune response to cancer, and represents a breakthrough in cancer treatment. Immune checkpoint inhibitors (ICIs) are a type of immunotherapy that consist of monoclonal antibodies that attempt to strengthen and revitalize the immune system by binding to coinhibitory receptors, prompting immune-mediated tumoral cell death (1). Because of their advantages in terms of therapeutic efficacy, including an unprecedented and durable antitumor response rate, ICIs have completely changed the therapeutic landscape for patients with a variety of cancer types. Currently, approved ICIs are directed against cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), programmed cell death protein-1 (PD-1), and programmed cell death ligand 1 (PD-L1). PD-1 and PD-L1 are coinhibitory proteins expressed by lymphocytes and antigen-presenting cells (APCs) that induce self-tolerance and autoimmunity regulation, while CTLA-4 is expressed in T cells and B cells and negatively regulate lymphocyte activation (2).
ICIs have shown excellent antitumor activity in a variety of cancers; however, ICIs can cause multiple organ damage and unique side effects called immune-related adverse events (irAEs). These irAEs affect virtually every organ system and can lead to significant morbidity, mortality and impaired quality of life. The systemic augmentation of immune responses by ICIs, especially when they are used in combination, can lead to a range of immune-related toxicities (3). Fortunately, these immune-mediated toxicities are largely reversible (4), but they require early, precise identification and timely intervention. One of the most common adverse events (AEs) is colitis, which can significantly lower a patient’s quality of life, and force antitumor medication to be stopped or halted. In clinical practice, the first-line treatment for irAE colitis is corticosteroids (CSs) (5). Further, biologics such as infliximab (IFX) and vedolizumab (VDZ) have gradually attracted attention, but they are often used as complementary treatments or as an upgrade after failure of the first-line treatment for irAE colitis.
The mechanism and effectiveness of first-line steroid therapy, and second-line biologics and other treatments for irAE colitis have not yet been fully summarized, and the pathogenesis and pathophysiology of irAE colitis remain unclear. Therefore, at present, there is no consensus as to when biologics should be used and which biologics should be administered. This review summarized the current mechanisms behind irAE colitis and its evidence-based management with an emphasis on the role of biological therapies to elucidate the mechanism, effectiveness, and safety of therapies for treating irAE colitis. Mechanism-based approaches will enable the application of appropriate treatment options for patients with irAE colitis, especially those with refractory irAE colitis. We present this article in accordance with the Narrative Review reporting checklist (available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-2150/rc).
Methods
A literature search was conducted of the PubMed database using the following keywords: “Immune checkpoint inhibitor-related colitis” AND “Corticosteroids” AND “Biologics” (Table 1). Only research articles written in English were considered, and no predefined restrictions were set in terms of the study type. The data sources were independently screened by two authors. The data analysis was conducted by two authors.
Table 1
Items | Specification |
---|---|
Dates of searches | 01 June 2024 to 01 August 2024 |
Database | PubMed |
Search terms | “Immune checkpoint inhibitor-related colitis” AND “Corticosteroids” AND “Biologics” |
Timeframe | 1995–2024 |
Inclusion and exclusion criteria | Inclusion criteria: all study types |
Exclusion criteria: articles published in languages other than English | |
Selection process | S.W. and H.W. independently screened the data sources. The data analysis was conducted by S.W. and H.W. |
Epidemiology
The lower gastrointestinal (GI) tract is the most commonly affected by ICI-related GI AEs. Colitis is defined as the presence of mucosal inflammation. It has been reported that the incidence of irAE colitis is 0.7–1.6% with anti-PD-1 therapy, 5.7–9.1% with anti-CTLA-4 therapy, and nearly 13.6% with the combination of anti-PD-1/PD-L1 and anti-CTLA-4 therapies (6-8). Overall, compared to anti-PD-1/PD-L1 therapy, anti-CTLA-4 therapy is more likely to result in immunological-related diarrhea and colitis. To date, there is no evidence that sex, tumor type, and the severity of immune-mediated colitis are significantly correlated (9). Colitis can develop any time after starting ICI medication, but occurs earlier with PD-1 inhibitors, and has a greater severity with anti-CTLA-4 therapy, and combination therapy in particular (9-12).
Mechanism
The exact mechanism of immune checkpoint inhibitor-associated colitis (ICIC) has not been defined; however, the current research results support the hypothesis that the proinflammatory response plays an important role in this process, causing a proinflammatory status and the emergence of an autoimmune-type presentation (13-15). According to Luoma et al., the overactivation of tissue-resident cluster of differentiation (CD)8+ T cells is a significant factor in colitis (16). Additional CD8+ and CD4+ T-cell populations are subsequently drawn from the blood on the activation of these T cells. Because a healthy colon already has a significant number of these tissue-resident CD8+ T cells, irAE colitis develops relatively early following the administration of ICIs. The development of ICIC is significantly influenced by the abundance of CD8 tissue-resident T cells in the colon, and their activation promotes the assembly of CD4 and CD8 T cells accompanied by the release of granzymes. CD8 tissue-resident memory T cells have been shown to be the dominant immune cell population in ICIC, and those located near the epithelial border drive its cytotoxicity (17). The blocking of immune checkpoints can achieve significant tumor regression in some patients; however, the systemic activation of autoreactive T cells damages off-target host tissues, causing a range of toxicities (4). Several animal models lacking immune checkpoints have been used to simulate the immunological effects of checkpoint inhibitor-associated colitis. Mice with the CTLA-4 deletion exhibit broad immune cell infiltration in numerous organs and lethal colitis due to enhanced T-cell activity (18-20).
Additionally, cytokines may play a part in ICIC. A preclinical model of ICIC has been shown to increase interleukin-17 (IL-17) (21,22), while inducible genes and the expression of interferon-γ (INF-γ) and tumor necrosis factor-α (TNF-α), which induce cell death, are substantially increased in ICIC patients (16,23). The expression of tumor necrosis factor-like cytokine 1A (TL1A) and its receptor death receptor 3 (DR3) is also upregulated in ICIC (24). In addition, the recombinant chemokine C-X-C-motif receptor 3 (CXCR3) and CXCR6 chemokine receptor (CXCR9/10 and CXCR16) genes are highly expressed in colitis-related T-cell populations, and increase T-cell activity (25). The development of irAE colitis may also be facilitated by the production of cytokines from cytotoxic T lymphocytes (CTLs) and the increased expression of chemokine receptor genes in colitis-associated T cells.
ICIs regulate microbiota-gut barrier homeostasis by causing apoptosis in intestinal epithelial cells, which disrupts barrier function. Pathogenic helper T 17 cells can further lead to an imbalance in the homeostasis of the gut microbiota resulting in severe intestinal toxicity, similar to the early signs of colitis (12). A negative link between anaerobic antibiotic treatment and the severity and prognosis of colitis has been shown recently, mirroring the importance of the gut microbiota and ecological dysbiosis in the pathogenesis of ICIC.
Clinical characteristics
ICIC has a variety of clinical symptoms. The most common symptom is diarrhea, and other symptoms include fever, abdominal discomfort, distention, mucus, blood in the stool, and peritoneal symptoms. Based on our clinical practice, as a symptom of ICIC, diarrhea is characterized by its frequent occurrence at night, and thus differs from diarrhea caused by other GI diseases. In most cases, ICIC appears quickly after the start of therapy, but it may also present later in the course of treatment (26-29). The diagnostic elements of ICIC include obtaining a complete and detailed history, ruling out the cause of infection, and assessing the degree of inflammation. Since many anticancer treatments also have GI side effects, it is critical to obtain a complete drug history in actual clinical studies and treatments. Because of the poor immune function of cancer patients, infection must be ruled out before diagnosis. The most common screening methods for infections are Clostridium difficile and cytomegalovirus tests. Colonoscopy is the gold standard for diagnosing ICIC, and is advised for patients with grade 2 or higher diarrhea. Endoscopic observations may vary from a nearly normal appearing mucosa to alterations such as mucosal erythema, loss of vascular pattern, edema, ulcerations, friability, and necrosis (30,31). Endoscopic and histological findings can sometimes differ, and normal mucosa can be found in roughly one-third of patients with ICIC (9). Thus, even if no clear evidence of inflammatory injury is observed endoscopically, biopsies of normal mucosa are necessary.
Biopsies are frequently used to aid in diagnosis; however, histopathological results are nonspecific (32). The main histological manifestations are neutrophilic infiltration, cryptitis, crypt abscesses, and chronic apoptosis changes such as intraepithelial lymphocytosis, crypt distortion, Paneth cell metaplasia, or basal plasmacytosis (33-35), which can also be seen in inflammatory bowel disease (IBD). Radiographic imaging is not required for the diagnosis of ICIC, but it can provide useful information, especially when there are no characteristic findings in the endoscopic evaluation. In some patients, computed tomography scans show bowel wall thickening, mesenteric vascular engorgement, fat stranding, and fluid-filled bowel (36).
When ICIC symptoms appear, their severity can be determined systematically using the Common Terminology Criteria for Adverse Events (CTCAE) (37): grade 1 irAE colitis causes asymptomatic or mild diarrhea; grade 2 colitis presents as mild abdominal pain, diarrhea, and hematochezia; grade 3 and grade 4 colitis are characterized by severe abdominal pain and frequent diarrhea, which can result in bleeding, intestinal obstruction, peritonitis, necrosis, and intestinal perforation; while Grade 5 colitis can be fatal (30,33,38,39). Diffuse enteritis occurs alone or in association with colitis in 25% of patients (40). While rare (0.3–1.3%), fatal AEs associated with ICIs primarily include colitis and toxic megacolon with colonic perforation, which are particularly common in anti-CTLA-4 therapy (41). Isolated upper GI inflammation (gastritis, gastroenteritis, or enteritis) occur with a frequency of more than 10% (13,42,43).
As the intestinal symptoms caused by ICIC are diverse, an accurate differential diagnosis is needed before a final diagnosis can be made, except for other etiologies, such as medications, infections, and IBD. The guidelines recommend stool testing for pathogens in patients who present with moderate to severe diarrhea, abdominal pain, fever, or GI bleeding after ICIC therapy. However, infectious factors may coexist with ICIC and common pathogens include cytomegalovirus, Clostridium difficile, Salmonella and Candida (12,44-46). ICIC and IBD have similar clinical and histologic features, and in some circumstances, their endoscopic and histological findings, such as patchy or segmental distribution and crypt architecture irregularities (40), are nearly superimposable (47). In addition to having similar clinical and endoscopic manifestations, ICIC and IBD share certain similarities in terms of pathogenesis. Both diseases increase regulatory cytokines (e.g., INF-γ and IL-17) at the mucosal level (24). Further, CTLA-4, PD-1/PD-L1, and the gut microbiome also play important roles in intestinal immunity in both IBD and ICIC patients (48). In a mouse model of IBD, PD-1 protein was found to be protective against colitis (22). In humans with Crohn’s disease (CD), intestinal APCs do not express PD-L1 (49). Conversely, several CTLA-4 polymorphisms are known to enhance the risk of developing both CD and ulcerative colitis (UC) (50). Given the above-mentioned similarities in the clinical manifestations, histology, immunology, and pathogenesis of ICIC and IBD, the current relatively mature treatment regimen for IBD may provide treatment options for ICIC.
Treatment
The treatments for ICI-related GI issues are tripartite and comprise: withholding ICIs to prevent further toxicity; inducing immunosuppression to reduce inflammatory changes; and providing supportive therapy to address GI complications. Treatment approaches vary greatly; however, it is widely agreed that ICIC therapy should be initiated, and ICI medication should be discontinued if the severity of symptoms exceed grade 2 (51). We discuss the clinical drugs used for the treatment of colitis, as well as their modes of action and mechanisms in the following sections.
CSs
CSs (prednisone 1–2 mg/kg/day or equivalent) are often the first line of treatment for grade 2 or above colitis (5). Additionally, given the characteristics of nocturnal diarrhea in ICIC patients, the divided administration of steroid hormones may be considered in clinical practice. CSs inhibit the development of dendritic cells and cause activated T cells to undergo apoptosis, suppressing both the innate and adaptive immune systems (52). Further, CSs also inhibit the production of proinflammatory cytokines such as IL-2 and IFN-γ from activated T cells (53). CSs have been shown to increase the surface expression of PD-1 in both CD4+ and CD8+ T cells and suppress their functions in mouse models (54). This suggests that systemic CS treatment could be used for ICICs. It should also be noted that the high-dose and long-term treatment of CSs raises the risk of side effects such as infections, osteoporosis, and decreased glucose tolerance (55), and recent research suggests that high-dose steroids may hinder optimal antitumor responses (56,57). The use of mesalazine and topical steroids as a therapeutic strategy in addition to systemic steroids to minimize the side effects of high-dose hormone use has also been reported
In addition, it is important to consider the patients with steroid-refractory or steroid-resistant ICIC. Steroid-refractory ICIC requires the use of immunosuppression due to an initial non-response to CSs, while steroid-resistant ICIC requires the use of immunosuppression after the initial response and the continued use of systemic steroids (58). In a series of retrospective studies of patients with ICI-related diarrhea/colitis (30,33,59), only approximately half of the patients treated with CSs experienced symptom relief. However, in some situations, CSs are unable to control symptoms, and in the absence of more aggressive management, diarrhea or colitis may continue, worsen, or even become life threatening. When symptoms improve, patients should be gradually weaned off CSs over 4 to 6 weeks (5); however, symptom recurrence may occur during the decrement process. Overall, 2% of patients require additional immunosuppressive therapies other than steroids, which may last for several months, and management is challenging.
Diarrhea and colitis are the most common irAEs (58). Approximately 30–60% of ICI-associated diarrhea/colitis patients have a tendency to be resistant to first-line CSs, showing no response to high-dose steroids within 72 hours or no complete remission within 1 week (30), which is related to the expression level of TNF-α. Given the function of TNF family proteins in response to steroid treatment, the following points cannot be ignored: (I) mucosal TNF-α expression levels are negatively correlated with susceptibility to CS (55); and (II) patients with CS-resistant irAE colitis exhibit elevated expression of IFN-γ signaling genes in their intestinal mucosa (30).
In summary, CS-refractory/resistant ICIC can cause serious conditions that can be fatal if treatment fails (58,60). Thus, further treatments are urgently needed. Due to the morphological and immunological similarities between ICIC and IBD, it has been proposed that biologic therapy be applied to ICIC. The management of CS-refractory/resistant irAEs is a growing challenge, and alternative treatment courses need to be explored. It is currently visible in the clinical practice of IBD, and endoscopic results should be taken into account to identify patients who should receive infliximab (IFX) or vedolizumab (VDZ), rather than waiting for a response to CSs. Current studies have shown that in addition to first-line hormone therapy, IFX and VDZ are the main treatments for steroid-refractory/resident ICIC patients (61,62).
IFX
IFX, a chimeric immunoglobulin G1 monoclonal antibody against TNF, causes immunosuppression. It is a TNF inhibitor that blocks the effects of the proinflammatory cytokine TNF-α. TNF-α signaling plays a significant role in cellular processes. It was first approved to treat UC and CD. In accordance with standard IBD therapies, IFX (5 mg/kg/dose) is administered intravenously to treat CS-resistant irAE colitis. For irAE colitis, the therapeutic response to IFX usually occurs in a few days, and symptoms improve with just one dose (63-65). Some patients need to take a second dose of IFX after two weeks. Evidence shows that the chance of endoscopic and histological remission can be increased, and the risk of recurrence can be reduced if up to three doses (at weeks 0, 2, and 6) are administered (66).
In certain retrospective cohort studies (67,68), patients with IFX-treated CS-refractory irAE colitis have been reported to have remission rates of 54% and 71.4%, respectively. Researchers compared the clinical outcomes of patients with irAE colitis treated with CSs alone to those who received IFX after beginning CS therapy (69). Patients who received IFX after CS therapy experienced a considerably shorter time to symptom relief, suggesting that early IFX administration should be considered. In most published case series, positive results have been reported with TNF-α inhibition, with almost all patients with colitis improving after a single dose of a TNF-α inhibitor. Thus, it is not only beneficial for symptom control but also for shorter hospital stays, which supports the strategy of the early initiation of TNF-α suppression rather than systemic steroid therapy that lasts for months. Notably, before the initiation of IFX, an infectious workup for human immunodeficiency virus (HIV), tuberculosis, and hepatitis should be performed to exclude latent infection.
VDZ
VDZ can be used in cases where IFX is contraindicated or ineffective, and is the first choice when anti-TNF agents are contraindicated. VDZ is an immunoglobulin G monoclonal antibody that binds to α4β7 integrin in activated T cells. By preventing memory T cells from sticking to and penetrating the intestinal wall (70), it also hinders the entry of activated T cells into intestinal tissue by blocking their interaction with mucosal address in cell adhesion molecule-1 (MAdCAM-1), which is specifically expressed in intestinal vascular endothelial cells (71,72). Abu-Sbeih et al. investigated the effects of VDZ on irAE colitis that was resistant to CSs and/or IFX (73). The remission rate of VDZ was 67% in patients who received IFX before, and 95% in patients who did not receive IFX, and no side effects were noted. The exact duration of VDZ has not been determined; however, similar to IFX, a study has found that the risk of recurrence will be reduced and the chance of endoscopic or histologic remission will be increased if up to three doses (at weeks 0, 2, and 6) are administered (23). Infectious testing, such as HIV and tuberculosis testing, should also be undertaken to exclude possible infection before VDZ treatment is initiated. Notably, VDZ is a gut-specific agent, and it is unclear whether VDZ can be used to treat other non-GI-related irAEs (74,75).
VDZ can specifically limit the interaction of leukocytes with the intestinal vasculature and prevent the influx of inflammatory cells, which mediate the inflammatory process in immune-related colitis (76). Because of its highly gut-selective mechanism, long-term VDZ treatment has a remarkable safety profile (77), and its administration in clinical practice may lead to fewer systemic immunosuppression and adverse effects. An observational study comparing VDZ and IFX in the treatment of ICIC reported similar response rates in patients treated with the two monoclonal antibodies, but the VDZ group had a longer duration of clinical remission. Moreover, VDZ was linked to shorter CS exposure. Further, compared to patients receiving IFX alone, those receiving VDZ monotherapy had better results and a decreased rate of cancer progression. A notable decrease in the infection rate was also detected in the VDZ group (75).
Both VDZ and IFX have similar efficacy, but the safety profile of VDZ is greater than that of IFX. Choosing between IFX and VDZ for CS-refractory patients is challenging due to limited data, but in general, IFX should be avoided in patients with confirmed or suspected infections, or other contraindications to TNF-α inhibitors, while extreme caution should be exercised when considering whether to administer VDZ to patients with GI tract cancers, metastasis, or GI tract infections (78). The use of VDZ is attractive in this patient population due to its safety profile, but IFX may achieve faster remission based on its efficacy in acute, hospitalized patients (79,80).
Other therapeutic agents
There are not enough well-established alternative therapy choices for patients who do not respond to normal standard care (81). New therapeutic agents for treating ICIC include tofacitinib, tocilizumab, ustekinumab, and mycophenolate mofetil (MMF), but these new therapeutic agents are currently available off-label. Using the JAK inhibitor tofacitinib, Sasson et al. successfully inhibited T-cell function in a patient with refractory ICIC (82). Tocilizumab is an antibody that works by binding to the IL-6 receptor to help lower the amount of proinflammatory cytokines produced (83). Tocilizumab can be safely used to treat ICIC, and has been reported a clinical benefit rate of 84%. It should be noted that clinical trials have reported intestinal perforation with tocilizumab, especially in patients with GI ulcers and long-term hormone therapy, where the risk of intestinal perforation is increased (84). Therefore, tocilizumab should be administered carefully in patients with a history of long-term steroid hormone use and those with ulcerative lesions.
Ustekinumab, a monoclonal antibody facilitates mucosal healing by blocking a crucial part of the inflammatory helper T-cell pathway. Two cases have reported that Ustekinumab was effective in the patients who were initially refractory to CSs, IFX, and VDZ (85). MMF inhibits inosine-50-monophosphate dehydrogenase (IMPDH) and exerts immunosuppressive effects by preventing T-cell and B-cell multiplication. Mir et al. examined 11 patients with irAE colitis who were treated with MMF in combination with CSs, and reported that seven of these patients had no recurrence during CS reduction, and the remaining patients responded strongly to IFX (86). Calcineurin inhibitors bind to calcineurin by forming an intracellular complex with FK506-binding protein 12, inhibiting the release of cytokines and exhibiting a robust immunosuppressive effect by inhibiting T-cell activation (87). Kunogi et al. reported a case in which symptoms improved following tacrolimus treatment for irAE colitis refractory to CSs, IFX, and VDZ (88).
The mechanisms of CSs, biologics, and other therapeutic agents for ICIC are shown in Figure 1.
In summary, CS-refractory/resident patients treated with IFX or VDZ had better remission rates. Therefore, the use of biologics (alone or in combination with CSs) should be explored early in the treatment of high-risk patients and not only as an escalation after a failure to respond to CSs. The early use of IFX or VDZ was significantly related to fewer hospitalizations and a shorter duration of steroid treatment. High-risk patients were chosen primarily based on the severity of the ICIC, response to CSs, and the development of large and deep mucosal ulcerations, as well as extensive inflammation beyond the left colon (66,89,90). There is growing evidence that endoscopy may not only confirm an ICIC diagnosis but also provide insights into the optimal immunosuppressive regimen (33,42,59,91). Mooradian et al. reported that the Mayo Endoscopic Score (MES) was significantly higher in patients who required IFX than those who did not, while those with a MES of zero rarely required further immunosuppression. They also found no correlation between the intensity of clinical symptoms and the MES. According to these findings, similar to IBD, endoscopic characteristics can inform clinical judgments more effectively than ICIC patient symptoms alone (92). For ICIC, endoscopy is useful for identifying both high-risk patients and those who may benefit from early biologic use.
In the past, biologics were usually used as second-line treatments after the failure of hormone therapy for ICIC. However, by summarizing the above related mechanisms of the possible occurrence of ICIC and current therapies involving CSs and biologics, we found that CSs and biologics have different targets and affect their respective signaling pathways. Therefore, biologics should not be considered as an alternative and complementary option after the failure of CS therapy, but should be considered as an early treatment. However, the risk of infection caused by the extensive immunosuppressive effects of hormones combined with biologics also requires attention. At present, it is still uncertain whether all patients will benefit from the early use of biologics; thus, the precise identification of patient groups that can benefit from early biologics treatment should receive more attention, and achieving optimal clinical outcomes through more precise and personalized treatment strategies is a challenge that should be addressed in the future.
Microbiome alteration and fecal microbiota transplantation
The gut microbiome has been extensively studied in IBD, and changes in microbiome patterns associated with inflammatory activity include a decrease in the abundance of microorganisms, including Mycobacterium anisopliae, Verrucomicrobiae, and Bifidobacteriaceae, and an increase in the abundance of Clostridiaceae, Enterobacteriaceae, Enterococci, and Streptococci (93-96). Similarly, the gut microbiome plays an important part in the process of ICIC. Recent studies have shown that certain bacteria are associated with the onset and severity of ICIC (97,98). The bacterial taxa in the ICIC are predominantly Clostridium difficile and Escherichia coli with a few anaplastic and warty microorganisms (99). Therefore, there is a great deal of overlap between the microbiome characteristics of ICIC patients and those of IBD patients. Gut microecological dysregulation can trigger intestinal inflammation (100,101), and the microbiome plays an important role in the tumor response to immunotherapy, and the risk of ICIC development (102).
Given the critical role that ecological dysbiosis plays in ICIC, fecal transplants have been considered as a potential treatment for specific patients. Fecal transplants can restore the normal gut microbiota and reduce mucosal inflammation. To date, international guidelines have recommended it for the treatment of recurrent C. difficile colitis, but there is insufficient clinical evidence about its use in the treatment of ICIC. There are some case reports of successful fecal transplantation for the treatment of refractory and severe ICIC; however, there are no relevant data on the timing of the treatment or complications (103,104). Thus, further studies need to be conducted in the future to provide more adequate evidence for its clinical use.
Conclusions
The use of ICIs has revolutionized the prospects of tumor therapy and improved the prognosis of many patients. However, ICIs can also lead to irAEs, the most common of which is ICIC, which can severely reduce the quality of life of patients, and result in the termination of immunotherapy. Biologics are currently recommended for the treatment of ICIC, but are usually used as a supplement after the failure of first-line CS therapy. Increasing data suggest that patients with irAE colitis respond favorably to biologics, and that patients with CS-resistant/refractory enterocolitis can benefit from the early use of biologics. Biologics (alone or in combination with CS) should be considered as an early therapy option for high-risk patients rather than just as an escalation after a failure to respond to CSs. In terms of the selection of biologics, IFX and VDZ have similar overall response rates, but IFX has a faster onset of action than VDZ. Conversely, VDZ can cause IFX resistance or have contraindications to its use, but it is safe due to its high intestinal selectivity and can be used as a prophylactic agent. However, early use of biologics is still in the clinical practice phase with limited retrospective studies. There will need more prospective research in the future. The management of patients with ICIC needs to be individualized and refined. The gut microbiota is an emerging area that can provide information about the mechanisms and potential therapeutic targets of ICIC.
Acknowledgments
Funding: None.
Footnote
Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-2150/rc
Peer Review File: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-2150/prf
Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-2150/coif). The authors have no conflicts of interest to declare.
Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.
References
- Raskov H, Orhan A, Christensen JP, et al. Cytotoxic CD8(+) T cells in cancer and cancer immunotherapy. Br J Cancer 2021;124:359-67. [Crossref] [PubMed]
- Bagchi S, Yuan R, Engleman EG. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu Rev Pathol 2021;16:223-49. [Crossref] [PubMed]
- Johnson DB, Chandra S, Sosman JA. Immune Checkpoint Inhibitor Toxicity in 2018. JAMA 2018;320:1702-3. [Crossref] [PubMed]
- Friedman CF, Proverbs-Singh TA, Postow MA. Treatment of the Immune-Related Adverse Effects of Immune Checkpoint Inhibitors: A Review. JAMA Oncol 2016;2:1346-53. [Crossref] [PubMed]
- Thompson JA, Schneider BJ, Brahmer J, et al. NCCN Guidelines Insights: Management of Immunotherapy-Related Toxicities, Version 1.2020. J Natl Compr Canc Netw 2020;18:230-41. [Crossref] [PubMed]
- Wang DY, Ye F, Zhao S, et al. Incidence of immune checkpoint inhibitor-related colitis in solid tumor patients: A systematic review and meta-analysis. Oncoimmunology 2017;6:e1344805. [Crossref] [PubMed]
- Tandon P, Bourassa-Blanchette S, Bishay K, et al. The Risk of Diarrhea and Colitis in Patients With Advanced Melanoma Undergoing Immune Checkpoint Inhibitor Therapy: A Systematic Review and Meta-Analysis. J Immunother 2018;41:101-8. [Crossref] [PubMed]
- Bishay K, Tandon P, Bourassa-Blanchette S, et al. The risk of diarrhea and colitis in patients with lung cancer treated with immune checkpoint inhibitors: a systematic review and meta-analysis. Curr Oncol 2020;27:e486-94. [Crossref] [PubMed]
- Abu-Sbeih H, Ali FS, Wang Y. Immune-checkpoint inhibitors induced diarrhea and colitis: a review of incidence, pathogenesis and management. Curr Opin Gastroenterol 2020;36:25-32. [Crossref] [PubMed]
- Arnaud-Coffin P, Maillet D, Gan HK, et al. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int J Cancer 2019;145:639-48. [Crossref] [PubMed]
- Tran AN, Wang M, Hundt M, et al. Immune Checkpoint Inhibitor-associated Diarrhea and Colitis: A Systematic Review and Meta-analysis of Observational Studies. J Immunother 2021;44:325-34. [Crossref] [PubMed]
- Tang L, Wang J, Lin N, et al. Immune Checkpoint Inhibitor-Associated Colitis: From Mechanism to Management. Front Immunol 2021;12:800879. [Crossref] [PubMed]
- Som A, Mandaliya R, Alsaadi D, et al. Immune checkpoint inhibitor-induced colitis: A comprehensive review. World J Clin Cases 2019;7:405-18. [Crossref] [PubMed]
- Liu YH, Zang XY, Wang JC, et al. Diagnosis and Management of Immune Related Adverse Events (irAEs) in Cancer Immunotherapy. Biomed Pharmacother 2019;120:109437. [Crossref] [PubMed]
- Marthey L, Mateus C, Mussini C, et al. Cancer Immunotherapy with Anti-CTLA-4 Monoclonal Antibodies Induces an Inflammatory Bowel Disease. J Crohns Colitis 2016;10:395-401. [Crossref] [PubMed]
- Luoma AM, Suo S, Williams HL, et al. Molecular Pathways of Colon Inflammation Induced by Cancer Immunotherapy. Cell 2020;182:655-671.e22. [Crossref] [PubMed]
- van Eijs MJM, Ter Linde JJM, Baars MJD, et al. Highly multiplexed spatial analysis identifies tissue-resident memory T cells as drivers of ulcerative and immune checkpoint inhibitor colitis. iScience 2023;26:107891. [Crossref] [PubMed]
- Wing K, Onishi Y, Prieto-Martin P, et al. CTLA-4 control over Foxp3+ regulatory T cell function. Science 2008;322:271-5. [Crossref] [PubMed]
- Tivol EA, Borriello F, Schweitzer AN, et al. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 1995;3:541-7. [Crossref] [PubMed]
- Klocke K, Sakaguchi S, Holmdahl R, et al. Induction of autoimmune disease by deletion of CTLA-4 in mice in adulthood. Proc Natl Acad Sci U S A 2016;113:E2383-92. [Crossref] [PubMed]
- Postow MA, Sidlow R, Hellmann MD. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N Engl J Med 2018;378:158-68. [Crossref] [PubMed]
- Song MY, Hong CP, Park SJ, et al. Protective effects of Fc-fused PD-L1 on two different animal models of colitis. Gut 2015;64:260-71. [Crossref] [PubMed]
- Badran YR, Cohen JV, Brastianos PK, et al. Concurrent therapy with immune checkpoint inhibitors and TNFα blockade in patients with gastrointestinal immune-related adverse events. J Immunother Cancer 2019;7:226. [Crossref] [PubMed]
- Bamias G, Delladetsima I, Perdiki M, et al. Immunological Characteristics of Colitis Associated with Anti-CTLA-4 Antibody Therapy. Cancer Invest 2017;35:443-55. [Crossref] [PubMed]
- Tokunaga R, Zhang W, Naseem M, et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation - A target for novel cancer therapy. Cancer Treat Rev 2018;63:40-7. [Crossref] [PubMed]
- Portenkirchner C, Kienle P, Horisberger K. Checkpoint Inhibitor-Induced Colitis-A Clinical Overview of Incidence, Prognostic Implications and Extension of Current Treatment Options. Pharmaceuticals (Basel) 2021;14:367. [Crossref] [PubMed]
- Marin-Acevedo JA, Harris DM, Burton MC. Immunotherapy-Induced Colitis: An Emerging Problem for the Hospitalist. J Hosp Med 2018;13:413-8. [Crossref] [PubMed]
- Couey MA, Bell RB, Patel AA, et al. Delayed immune-related events (DIRE) after discontinuation of immunotherapy: diagnostic hazard of autoimmunity at a distance. J Immunother Cancer 2019;7:165. [Crossref] [PubMed]
- Sarofim M, Winn R. Rare case of delayed onset colitis due to immunotherapy for malignant melanoma. ANZ J Surg 2019;89:E472-3. [Crossref] [PubMed]
- Geukes Foppen MH, Rozeman EA, van Wilpe S, et al. Immune checkpoint inhibition-related colitis: symptoms, endoscopic features, histology and response to management. ESMO Open 2018;3:e000278. [Crossref] [PubMed]
- Nishida T, Iijima H, Adachi S. Immune checkpoint inhibitor-induced diarrhea/colitis: Endoscopic and pathologic findings. World J Gastrointest Pathophysiol 2019;10:17-28. [Crossref] [PubMed]
- Grover S, Srivastava A. Lymphocytic colitis-like pattern of mucosal injury and the challenges in diagnosing cancer immunotherapy-related toxicity. Cancer 2019;125:1768-70. [Crossref] [PubMed]
- Wang Y, Abu-Sbeih H, Mao E, et al. Endoscopic and Histologic Features of Immune Checkpoint Inhibitor-Related Colitis. Inflamm Bowel Dis 2018;24:1695-705. [Crossref] [PubMed]
- Chen JH, Pezhouh MK, Lauwers GY, et al. Histopathologic Features of Colitis Due to Immunotherapy With Anti-PD-1 Antibodies. Am J Surg Pathol 2017;41:643-54. [Crossref] [PubMed]
- Verschuren EC, van den Eertwegh AJ, Wonders J, et al. Clinical, Endoscopic, and Histologic Characteristics of Ipilimumab-Associated Colitis. Clin Gastroenterol Hepatol 2016;14:836-42. [Crossref] [PubMed]
- Shieh AC, Guler E, Pfau D, et al. Imaging and clinical manifestations of immune checkpoint inhibitor-related colitis in cancer patients treated with monotherapy or combination therapy. Abdom Radiol (NY) 2020;45:3028-35. [Crossref] [PubMed]
- Mendoza TR, Dueck AC, Bennett AV, et al. Evaluation of different recall periods for the US National Cancer Institute's PRO-CTCAE. Clin Trials 2017;14:255-63. [Crossref] [PubMed]
- Beck KE, Blansfield JA, Tran KQ, et al. Enterocolitis in patients with cancer after antibody blockade of cytotoxic T-lymphocyte-associated antigen 4. J Clin Oncol 2006;24:2283-9. [Crossref] [PubMed]
- Wang DY, Mooradian MJ, Kim D, et al. Clinical characterization of colitis arising from anti-PD-1 based therapy. Oncoimmunology 2018;8:e1524695. [Crossref] [PubMed]
- Martins F, Sofiya L, Sykiotis GP, et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol 2019;16:563-80. [Crossref] [PubMed]
- Wang DY, Salem JE, Cohen JV, et al. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol 2018;4:1721-8. [Crossref] [PubMed]
- Hughes MS, Molina GE, Chen ST, et al. Budesonide treatment for microscopic colitis from immune checkpoint inhibitors. J Immunother Cancer 2019;7:292. [Crossref] [PubMed]
- Haryal A, Townsend MJ, Baskaran V, et al. Immune checkpoint inhibitor gastritis is often associated with concomitant enterocolitis, which impacts the clinical course. Cancer 2023;129:367-75. [Crossref] [PubMed]
- Ni J, Zhang X, Zhang L. Opportunistic bowel infection after corticosteroid dosage tapering in a stage IV lung cancer patient with tislelizumab-related colitis. Thorac Cancer 2020;11:1699-702. [Crossref] [PubMed]
- Zhou C, Klionsky Y, Treasure ME, et al. Pembrolizumab-Induced Immune-Mediated Colitis in a Patient with Concurrent Clostridium Difficile Infection. Case Rep Oncol 2019;12:164-70. [Crossref] [PubMed]
- Vuillamy C, Arnault JP, Fumery M, et al. Clostridium difficile infection and immune checkpoint inhibitor-induced colitis in melanoma: 18 cases and a review of the literature. Melanoma Res 2023;33:192-8. [Crossref] [PubMed]
- Del Sordo R, Lougaris V, Bassotti G, et al. Therapeutic agents affecting the immune system and drug-induced inflammatory bowel disease (IBD): A review on etiological and pathogenetic aspects. Clin Immunol 2022;234:108916. [Crossref] [PubMed]
- Shirwaikar Thomas A, Hanauer S, Wang Y. Immune Checkpoint Inhibitor Enterocolitis vs Idiopathic Inflammatory Bowel Disease. Clin Gastroenterol Hepatol 2023;21:878-90. [Crossref] [PubMed]
- Robertson J, Haas CT, Pele LC, et al. Intestinal APCs of the endogenous nanomineral pathway fail to express PD-L1 in Crohn's disease. Sci Rep 2016;6:26747. [Crossref] [PubMed]
- Zhang M, Ni J, Xu WD, et al. Association of CTLA-4 variants with susceptibility to inflammatory bowel disease: a meta-analysis. Hum Immunol 2014;75:227-33. [Crossref] [PubMed]
- Allouchery M, Beuvon C, Pérault-Pochat MC, et al. Safety of Immune Checkpoint Inhibitor Resumption after Interruption for Immune-Related Adverse Events, a Narrative Review. Cancers (Basel) 2022;14:955. [Crossref] [PubMed]
- Giles AJ, Hutchinson MND, Sonnemann HM, et al. Dexamethasone-induced immunosuppression: mechanisms and implications for immunotherapy. J Immunother Cancer 2018;6:51. [Crossref] [PubMed]
- Almawi WY, Beyhum HN, Rahme AA, et al. Regulation of cytokine and cytokine receptor expression by glucocorticoids. J Leukoc Biol 1996;60:563-72. [Crossref] [PubMed]
- Xing K, Gu B, Zhang P, et al. Dexamethasone enhances programmed cell death 1 (PD-1) expression during T cell activation: an insight into the optimum application of glucocorticoids in anti-cancer therapy. BMC Immunol 2015;16:39. [Crossref] [PubMed]
- Stone ML, Forster EM. Use of Vedolizumab in Immune Checkpoint Inhibitor-associated Enterocolitis. Inflamm Bowel Dis 2021;27:e147. [Crossref] [PubMed]
- Faje AT, Lawrence D, Flaherty K, et al. High-dose glucocorticoids for the treatment of ipilimumab-induced hypophysitis is associated with reduced survival in patients with melanoma. Cancer 2018;124:3706-14. [Crossref] [PubMed]
- Arbour KC, Mezquita L, Long N, et al. Impact of Baseline Steroids on Efficacy of Programmed Cell Death-1 and Programmed Death-Ligand 1 Blockade in Patients With Non-Small-Cell Lung Cancer. J Clin Oncol 2018;36:2872-8. [Crossref] [PubMed]
- Luo J, Beattie JA, Fuentes P, et al. Beyond Steroids: Immunosuppressants in Steroid-Refractory or Resistant Immune-Related Adverse Events. J Thorac Oncol 2021;16:1759-64. [Crossref] [PubMed]
- Jain A, Lipson EJ, Sharfman WH, et al. Colonic ulcerations may predict steroid-refractory course in patients with ipilimumab-mediated enterocolitis. World J Gastroenterol 2017;23:2023-8. [Crossref] [PubMed]
- Abdel-Wahab N, Shah M, Suarez-Almazor ME. Adverse Events Associated with Immune Checkpoint Blockade in Patients with Cancer: A Systematic Review of Case Reports. PLoS One 2016;11:e0160221. [Crossref] [PubMed]
- Soularue E, Lepage P, Colombel JF, et al. Enterocolitis due to immune checkpoint inhibitors: a systematic review. Gut 2018;67:2056-67. [Crossref] [PubMed]
- Haanen JBAG, Carbonnel F, Robert C, et al. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2017;28:iv119-42. [Crossref] [PubMed]
- Yanai S, Nakamura S, Matsumoto T. Nivolumab-Induced Colitis Treated by Infliximab. Clin Gastroenterol Hepatol 2017;15:e80-1. [Crossref] [PubMed]
- Callens R, Tamsin A, van Zandweghe L. Nivolumab-Induced Fulminant Immune-Related Colitis Despite Infliximab in a Patient With NSCLC. J Thorac Oncol 2019;14:e49-50. [Crossref] [PubMed]
- Paparoupa M, Stupperich S, Goerg-Reifenberg L, et al. Successful Treatment of an Immune-Mediated Colitis Induced by Checkpoint Inhibitor Therapy in a Patient with Advanced Melanoma. Case Rep Gastroenterol 2020;14:554-60. [Crossref] [PubMed]
- Abu-Sbeih H, Ali FS, Wang X, et al. Early introduction of selective immunosuppressive therapy associated with favorable clinical outcomes in patients with immune checkpoint inhibitor-induced colitis. J Immunother Cancer 2019;7:93. [Crossref] [PubMed]
- Hillock NT, Heard S, Kichenadasse G, et al. Infliximab for ipilimumab-induced colitis: A series of 13 patients. Asia Pac J Clin Oncol 2017;13:e284-90. [Crossref] [PubMed]
- Alexander JL, Ibraheim H, Sheth B, et al. Clinical outcomes of patients with corticosteroid refractory immune checkpoint inhibitor-induced enterocolitis treated with infliximab. J Immunother Cancer 2021;9:e002742. [Crossref] [PubMed]
- Johnson DH, Zobniw CM, Trinh VA, et al. Infliximab associated with faster symptom resolution compared with corticosteroids alone for the management of immune-related enterocolitis. J Immunother Cancer 2018;6:103. [Crossref] [PubMed]
- Klemm N, Trasolini R, Chahal D, et al. Vedolizumab for steroid & infliximab refractory immune checkpoint inhibitor associated colitis. Clin Res Hepatol Gastroenterol 2021;45:101604. [Crossref] [PubMed]
- Sandborn WJ, Feagan BG, Rutgeerts P, et al. Vedolizumab as induction and maintenance therapy for Crohn's disease. N Engl J Med 2013;369:711-21. [Crossref] [PubMed]
- Feagan BG, Rutgeerts P, Sands BE, et al. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2013;369:699-710. [Crossref] [PubMed]
- Abu-Sbeih H, Ali FS, Alsaadi D, et al. Outcomes of vedolizumab therapy in patients with immune checkpoint inhibitor-induced colitis: a multi-center study. J Immunother Cancer 2018;6:142. [Crossref] [PubMed]
- Kaneoka A, Okada E, Sugino H, et al. Vedolizumab Attenuates Immune-Checkpoint-Therapy-Induced Infliximab-Refractory Colitis. Diagnostics (Basel) 2022;12:480. [Crossref] [PubMed]
- Zou F, Faleck D, Thomas A, et al. Efficacy and safety of vedolizumab and infliximab treatment for immune-mediated diarrhea and colitis in patients with cancer: a two-center observational study. J Immunother Cancer 2021;9:e003277. [Crossref] [PubMed]
- Biemans VBC, Hoentjen F, Pierik MJ. Vedolizumab versus Adalimumab for Moderate-to-Severe Ulcerative Colitis. N Engl J Med 2020;382:92. [Crossref] [PubMed]
- Loftus EV Jr, Colombel JF, Feagan BG, et al. Long-term Efficacy of Vedolizumab for Ulcerative Colitis. J Crohns Colitis 2017;11:400-11. [PubMed]
- Dougan M. Gastrointestinal and Hepatic Complications of Immunotherapy: Current Management and Future Perspectives. Curr Gastroenterol Rep 2020;22:15. [Crossref] [PubMed]
- Probert CS, Hearing SD, Schreiber S, et al. Infliximab in moderately severe glucocorticoid resistant ulcerative colitis: a randomised controlled trial. Gut 2003;52:998-1002. [Crossref] [PubMed]
- Järnerot G, Hertervig E, Friis-Liby I, et al. Infliximab as rescue therapy in severe to moderately severe ulcerative colitis: a randomized, placebo-controlled study. Gastroenterology 2005;128:1805-11. [Crossref] [PubMed]
- Dougan M, Wang Y, Rubio-Tapia A, et al. AGA Clinical Practice Update on Diagnosis and Management of Immune Checkpoint Inhibitor Colitis and Hepatitis: Expert Review. Gastroenterology 2021;160:1384-93. [Crossref] [PubMed]
- Sasson SC, Slevin SM, Cheung VTF, et al. Interferon-Gamma-Producing CD8(+) Tissue Resident Memory T Cells Are a Targetable Hallmark of Immune Checkpoint Inhibitor-Colitis. Gastroenterology 2021;161:1229-1244.e9. [Crossref] [PubMed]
- Holmstroem RB, Nielsen OH, Jacobsen S, et al. COLAR: open-label clinical study of IL-6 blockade with tocilizumab for the treatment of immune checkpoint inhibitor-induced colitis and arthritis. J Immunother Cancer 2022;10:e005111. [Crossref] [PubMed]
- Gout T, Ostör AJ, Nisar MK. Lower gastrointestinal perforation in rheumatoid arthritis patients treated with conventional DMARDs or tocilizumab: a systematic literature review. Clin Rheumatol 2011;30:1471-4. [Crossref] [PubMed]
- Thomas AS, Ma W, Wang Y. Ustekinumab for Refractory Colitis Associated with Immune Checkpoint Inhibitors. N Engl J Med 2021;384:581-3. [Crossref] [PubMed]
- Mir R, Shaw HM, Nathan PD. Mycophenolate mofetil alongside high-dose corticosteroids: optimizing the management of combination immune checkpoint inhibitor-induced colitis. Melanoma Res 2019;29:102-6. [Crossref] [PubMed]
- Ohwada S, Ishigami K, Akutsu N, et al. Pharmacological Treatments Available for Immune-Checkpoint-Inhibitor-Induced Colitis. Biomedicines 2022;10:1334. [Crossref] [PubMed]
- Kunogi Y, Tominaga K, Abe K, et al. Refractory Immune Checkpoint Inhibitor-Induced Colitis Improved by Tacrolimus: A Case Report. Healthcare (Basel) 2021;9:418. [Crossref] [PubMed]
- Abu-Sbeih H, Ali FS, Luo W, et al. Importance of endoscopic and histological evaluation in the management of immune checkpoint inhibitor-induced colitis. J Immunother Cancer 2018;6:95. [Crossref] [PubMed]
- Gong Z, Wang Y. Immune Checkpoint Inhibitor-Mediated Diarrhea and Colitis: A Clinical Review. JCO Oncol Pract 2020;16:453-61. [Crossref] [PubMed]
- Geukes Foppen MH, Boogerd W, Blank CU, et al. Clinical and radiological response of BRAF inhibition and MEK inhibition in patients with brain metastases from BRAF-mutated melanoma. Melanoma Res 2018;28:126-33. [Crossref] [PubMed]
- Mooradian MJ, Wang DY, Coromilas A, et al. Mucosal inflammation predicts response to systemic steroids in immune checkpoint inhibitor colitis. J Immunother Cancer 2020;8:e000451. [Crossref] [PubMed]
- Rehman A, Rausch P, Wang J, et al. Geographical patterns of the standing and active human gut microbiome in health and IBD. Gut 2016;65:238-48. [Crossref] [PubMed]
- Alipour M, Zaidi D, Valcheva R, et al. Mucosal Barrier Depletion and Loss of Bacterial Diversity are Primary Abnormalities in Paediatric Ulcerative Colitis. J Crohns Colitis 2016;10:462-71. [Crossref] [PubMed]
- Eun CS, Kwak MJ, Han DS, et al. Does the intestinal microbial community of Korean Crohn's disease patients differ from that of western patients? BMC Gastroenterol 2016;16:28. [Crossref] [PubMed]
- Mar JS, LaMere BJ, Lin DL, et al. Disease Severity and Immune Activity Relate to Distinct Interkingdom Gut Microbiome States in Ethnically Distinct Ulcerative Colitis Patients. mBio 2016;7:e01072-16. [Crossref] [PubMed]
- Dubin K, Callahan MK, Ren B, et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun 2016;7:10391. [Crossref] [PubMed]
- Chaput N, Lepage P, Coutzac C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol 2017;28:1368-79. [Crossref] [PubMed]
- Png CW, Lindén SK, Gilshenan KS, et al. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am J Gastroenterol 2010;105:2420-8. [Crossref] [PubMed]
- Chassaing B, Darfeuille-Michaud A. The commensal microbiota and enteropathogens in the pathogenesis of inflammatory bowel diseases. Gastroenterology 2011;140:1720-28. [Crossref] [PubMed]
- Lepage P, Häsler R, Spehlmann ME, et al. Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Gastroenterology 2011;141:227-36. [Crossref] [PubMed]
- Vétizou M, Pitt JM, Daillère R, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015;350:1079-84. [Crossref] [PubMed]
- Chen M, Liu M, Li C, et al. Fecal Microbiota Transplantation Effectively Cures a Patient With Severe Bleeding Immune Checkpoint Inhibitor-Associated Colitis and a Short Review. Front Oncol 2022;12:913217. [Crossref] [PubMed]
- Wang Y, Wiesnoski DH, Helmink BA, et al. Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis. Nat Med 2018;24:1804-8. [Crossref] [PubMed]