Abstract
Free full text
Fecal Microbiota Transplantation for refractory immune checkpoint inhibitor-associated colitis
Abstract
We report the first case series of ICI-associated colitis successfully treated with fecal microbiota transplantation (FMT), with reconstitution of the gut microbiome and a relative increase in the proportion of regulatory T cells (Tregs) within the colonic mucosa. These preliminary data provide evidence that modulation of the gut microbiome may abrogate ICI-associated colitis.
Immunotherapy has transformed the field of oncology improving long-term survival in patients across numerous cancer types. Treatments with immune checkpoint inhibitors (ICI) targeting cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell death protein 1 (PD-1) and programmed cell death ligand 1 (PD-L1) are associated with increased T cell activation and effective anti-tumor immune responses in a subset of patients, but treatment can be associated with serious immune-related adverse effects (irAEs) in some patients1. One of the most common toxicities is ICI-associated colitis. This can be quite severe and closely resembles colitis associated with autoimmune pathophysiology including inflammatory bowel disease (IBD)2,3. ICI-associated colitis is routinely treated with immunosuppressive therapy, including corticosteroids and/or agents targeting tumor necrosis factor alpha (TNF-alpha), all of which have significant side effects. Recommendations regarding optimal management of ICI-induced colitis continue to evolve4–9. Interestingly, recent randomized clinical studies suggest that other types of colitis, including Clostridium difficile-associated colitis and IBD, can be successfully treated by modulation of the gut microbiome with fecal microbiota transplantation (FMT)10.
Previously, we demonstrated that differential bacterial signatures comprise the gut microbiome of responders versus non-responders to ICI therapy and additionally showed that modulation of the gut microbiome of germ-free mice via FMT from patients could alter anti-tumor immunity and response to ICI therapy in gnotobiotic mice11–13. Other groups have also demonstrated differential bacterial signatures in the gut microbiome of patients who develop ICI-associated colitis14–16 and have shown that targeting specific bacterial taxa may abrogate ICI-related toxicity in pre-clinical models17. However, approaches incorporating modulation of the microbiome to treat ICI-associated colitis have not yet been explored in patients. Herein, we document results from the first reported case series of two patients with refractory ICI-associated colitis successfully treated with FMT and describe changes in the immune microenvironment following FMT, a potential mechanism of action through which modulation of the gut microbiome may impact ICI-associated colitis.
We sought to determine the impact of treatment with FMT from healthy donors in patients with refractory ICI-associated colitis and enrolled two patients onto this treatment protocol between June 2017 and January 2018 (CIND17–0036, CIND17–0058). Clinical courses for both patients are further detailed in Supplemental Figs. 1–3. The first patient was a 50-year-old female with high-grade metastatic urothelial carcinoma refractory to standard chemotherapy who was enrolled onto a trial of combined CTLA-4 and PD-1 blockade (NCT1928394). Two weeks after treatment initiation, she was hospitalized with CTCAE Grade ≥ 2 diarrhea/colitis. An infectious workup including PCR-based multiplex assay for common GI pathogens was negative, and colonoscopy demonstrated severe colitis that endoscopically resembled ulcerative colitis (Fig. 1a and Supplemental Figs. 3a and 4a). She received systemic corticosteroids, followed later by two doses of an anti-TNF-alpha agent (infliximab) as well as one dose of anti-integrin therapy (vedolizumab), but her symptoms persisted. She then received a single dose of FMT (50 grams of donor stool) via colonoscopy. The second patient enrolled was a 78-year-old male with prostate cancer refractory to chemotherapy and hormonal therapy who received two doses of ipilimumab in the context of a clinical trial (NCT02113657). Three months after treatment initiation, he was hospitalized with fever and CTCAE Grade ≥ 2 diarrhea/colitis. Infectious etiologies were excluded and colonoscopy confirmed the diagnosis of ICI-associated colitis, though with a Crohn’s colitis-like presentation (Fig. 1d and Supplemental Figs. 3b and 4b). His symptoms persisted despite systemic corticosteroids, infliximab and vedolizumab. He received two doses of FMT. The source of all three FMT products was from a single healthy unrelated donor, collected at three different time points.
Both patients had complete resolution of clinical symptoms following treatment with FMT, with eventual return to normal solid daily bowel movements without further bleeding (Supplemental Fig. 3). In the first patient, complete resolution occurred gradually within 2 weeks and she was weaned off steroids in 7 days (Supplemental Fig. 3a), while the second patient experienced partial improvement of GI symptoms but with persistent ulcers on follow-up colonoscopy and recurrent abdominal pain; he experienced complete resolution after a second FMT treatment (Supplemental Fig. 3b).
Endoscopic evaluation demonstrated significant mucosal inflammation and ulceration in both patients near the time of diagnosis of ICI-colitis, without substantial improvement after systemic corticosteroids, anti-TNF and anti-integrin agents. Following FMT, marked improvement was evident on endoscopic evaluation, with reduced inflammation and resolution of ulcerations (Fig. 1a and 1d and Supplemental Figs. 3 and 4). In the first patient, analysis of immune infiltrates in the colonic mucosa demonstrated a dense inflammatory infiltrate prior to FMT with a high density of CD8+ cytotoxic T lymphocytes and a low density of CD4+ FoxP3+ T cells (Fig. 1b-c and Supplemental Figs. 5a and 6a), consistent with findings from reports of autoimmune colitis18,19. Following FMT, there was a substantial reduction in CD8+ T-cell density with a concomitant increase in CD4+ FoxP3+ (Fig. 1b-c and Supplemental Figs. 5a and 6a), offering a potential mechanism through which FMT could abrogate ICI-associated toxicity. In the second patient, the density of all T cell subtypes analyzed decreased following FMT, but the CD4+ T cell population was relatively spared compared to the CD8+ T cell population, with persistence again noted for CD4+ and FoxP3+ cells (Figs. 1e-f and Supplemental Figs. 5b, 6b and 7).
Importantly, stool samples were collected from patients prior to FMT and serially following FMT to assess the diversity and composition of the microbiome before and after intervention via 16S sequencing (Supplemental Figs. 1–2). Results demonstrated variable pre-treatment alpha diversity compared to the healthy FMT donor (Fig. 2a). There was no observable trend in alpha diversity following treatment, measured by the inverse Simpson’s index. Numbers of observed OTUs, however, did consistently rise following each FMT in both patients, though in both cases they would later come back down (Fig. 2a). Principal coordinates analyses of unweighted UniFrac distances demonstrated that the gut microbiome of patients became most similar to the FMT donor immediately following treatment, but, in both cases later deviated away though remaining distinct compared to pre-FMT (Fig. 2b). An evaluation of unique 16S sequences20 detectable in patient pre-FMT baseline samples and FMT products was used to determine probable bacterial origins in longitudinal samples, demonstrating considerable abundances of bacteria of donor origin that stably joined the patient’s intestinal microbiome with each FMT procedure (Fig. 2c).
We next assessed bacterial taxa present at time of colitis in these patients as well as compositional changes in the gut microbiome following treatment with FMT. Bacterial taxa present at time of colitis were quite disparate between the two patients, with a predominance of Clostridia and a notable absence of bacteria shown to be protective against ICI-associated colitis and IBD such as Bacteroidia14,16 and Verrucomicrobiae21 respectively in the first patient and a predominance of Gammaproteobacteria (predominantly Escherichia) in the second patient, which is commonly seen in perturbed intestinal states22. Immediately following FMT in the first patient, donor FMT-derived bacteria had effectively colonized the intestinal tract, with nearly 75% of the sequences uniquely attributable to the FMT donor microbiome and a notably higher abundance of Akkermansia (Fig. 2d-e). By week 7 after FMT, Akkermansia now only accounted for a small portion of her microbiome and there was further expansion of Clostridia which were largely patient-derived in origin (Fig. 2d, left and and2e,2e, left). Of note, the patient also showed an expansion of Bifidobacterium after FMT, which recently was reported to abrogate ICI-related toxicity in a murine model17 (Fig. 2d, left and and2e,2e, left). In the second patient, there was a notable increase in the abundance of Blautia and Bifidobacterium species after FMT, which have been associated with reduced intestinal inflammation (Fig. 2d, right and and2e,2e, right)23,17. In addition to this, he had a decrease in the abundance of potentially pathogenic Escherichia and an increase in potentially beneficial Bacteroides after his first FMT (Fig. 2d, right and and2e,2e, right). After his second FMT, he had a higher abundance of Escherichia and eventual decrease in Bacteroides; however, his gastrointestinal symptoms steadily continued to improve (Fig. 2d, right and and2e,2e, right).
Together, these cases provide provocative and novel evidence that modulation of the gut microbiome via FMT can be associated with significant and rapid improvement of refractory ICI-associated colitis with early insights into potential mechanisms; however, significant limitations exist with this very small cohort and additional studies are needed to assess the utility of this approach as well as to provide further mechanistic insight. Given the widespread application of ICI with expanding indications across cancer types, it is anticipated that there will be an increased incidence in ICI-associated colitis and other irAEs. Thus, further investigations building on this approach are critically needed.
Acknowledgements of Funding Support:
Andrew Sabin Family Fellows Program (private donation): J.A.W.
MD Anderson Cancer Center’s Melanoma Moon Shot Program (710499-80-111538-19): J.A.W.
American Association for Cancer Research Stand Up to Cancer (SU2C-AACR-IRG-19-17): J.A.W.
National Institutes of Health (R01 CA219896-01A1): J.A.W.
National Institutes of Health (R01 HL124112): R.R.J.
Cancer Prevention and Research Institute of Texas Grant (RR160089): R.R.J.
Footnotes
Competing Interests Declaration:
J.A.W and V.G. are inventors on a US patent application (PCT/US17/53,717) submitted by The University of Texas MD Anderson Cancer Center that covers methods to enhance checkpoint blockade therapy by the microbiome. J.A.W. is a clinical and scientific advisor at Microbiome DX and a consultant at Biothera Pharma, Merck Sharp, and Dohme. J.A.W. has honoraria from speakers’ bureau of Dava Oncology, Bristol-Myers Squibb, Gilead, Illumina, Omniprex, Imedex and is an advisory board member for GlaxoSmithKline, Novartis, and Roche/Genentech, Astra-Zeneca. V.G. is a consultant at Microbiome DX, and reports honoraria from ExpertConnect. R.R.J. is on the scientific advisory board for Seres Therapeutics, Inc., has consulted for Ziopharm Oncology and Microbiome Dx, and holds patents licensed to Seres Therapeutics, Inc. M.T.T. serves on the advisory board for Novartis, Seattle Genetics and Myriad Genetics. J.A.W., P.S., and J.P.A. are members of the Parker Institute for Cancer Immunotherapy at MD Anderson Cancer Center. P.S. is a consultant for Bristol-Myers Squibb, Jounce Therapeutics, Helsinn, and GlaxoSmithKline and is also a stockholder from Jounce Therapeutics. J.P.A. is a consultant and stockholder for Jounce Therapeutics, receives royalties from Bristol-Myers Squibb, and has intellectual property with Bristol-Myers Squibb and Merck. The other authors declare no competing interests.
Data Availability Statement:
Patient-related data not included in the paper were generated as part of clinical trials and may be subject to patient confidentiality. Any data and materials that can be shared will be released via a Material Transfer Agreement. Sequence data has been deposited at the European Genome-phenome Archive (EGA), which is hosted by the EBI and the CRG, under accession number EGAS00001003217.
References
Citations & impact
Impact metrics
Article citations
Cold and hot tumors: from molecular mechanisms to targeted therapy.
Signal Transduct Target Ther, 9(1):274, 18 Oct 2024
Cited by: 0 articles | PMID: 39420203 | PMCID: PMC11491057
Review Free full text in Europe PMC
Immune checkpoint inhibitor-induced diarrhea and colitis: an overview.
Support Care Cancer, 32(10):680, 23 Sep 2024
Cited by: 0 articles | PMID: 39311981 | PMCID: PMC11420271
Review Free full text in Europe PMC
Editorial for the Special Issue "Gut Dysbiosis: Molecular Mechanisms and Therapies 2.0".
Biomedicines, 12(10):2186, 26 Sep 2024
Cited by: 0 articles | PMID: 39457499 | PMCID: PMC11504651
The Impact of Infections in Patients Treated with Atezolizumab Plus Bevacizumab for Unresectable Hepatocellular Carcinoma.
J Clin Med, 13(17):4994, 23 Aug 2024
Cited by: 0 articles | PMID: 39274206 | PMCID: PMC11396642
Pharmacomicrobiomics in precision cancer therapy: bench to bedside.
Front Immunol, 15:1428420, 09 Sep 2024
Cited by: 0 articles | PMID: 39315107 | PMCID: PMC11416994
Review Free full text in Europe PMC
Go to all (338) article citations
Data
Data behind the article
This data has been text mined from the article, or deposited into data resources.
BioStudies: supplemental material and supporting data
Clinical Trials
- (1 citation) ClinicalTrials.gov - NCT02113657
European Genome-Phenome Archive
- (1 citation) European Genome-Phenome Archive - EGAS00001003217
Similar Articles
To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.
Transferable Immunoglobulin A-Coated Odoribacter splanchnicus in Responders to Fecal Microbiota Transplantation for Ulcerative Colitis Limits Colonic Inflammation.
Gastroenterology, 162(1):166-178, 02 Oct 2021
Cited by: 46 articles | PMID: 34606847 | PMCID: PMC8678328
Single-cell transcriptomic analyses reveal distinct immune cell contributions to epithelial barrier dysfunction in checkpoint inhibitor colitis.
Nat Med, 30(5):1349-1362, 09 May 2024
Cited by: 1 article | PMID: 38724705
Beneficial Effects of Fecal Microbiota Transplantation on Ulcerative Colitis in Mice.
Dig Dis Sci, 61(8):2262-2271, 05 Feb 2016
Cited by: 43 articles | PMID: 26846120
Checkpoint Inhibitor-Induced Colitis.
Am J Gastroenterol, 115(2):202-210, 01 Feb 2020
Cited by: 43 articles | PMID: 31922959
Review
Funding
Funders who supported this work.
NCI NIH HHS (2)
Grant ID: R01 CA219896
Grant ID: T32 CA009599
NHLBI NIH HHS (1)
Grant ID: R01 HL124112