Introduction
Fecal microbiota transplant (FMT) hasbeen commonlyused to treat recurrent Clostridium difficile infections (rCDI), a hospital-acquired infectiontypically treated with prolonged courses of antibiotics, which maintain and exacerbate intestinal dysbiosis [1 ]. FMT restores normal gut ecology, and in this context is well-tolerated and efficacious with cure rates of 85% in rCDI [2–5 ].
Cancer immunotherapy, particularly immune-checkpoint inhibitors (ICI) targeting cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death 1 (PD-1) inhibitory immune checkpoints, has transformed the management of advanced cancers. In addition to tumor-intrinsic mechanisms mediating response and resistance to ICI, the role of the gut microbiome as a tumor-extrinsic regulator of responses to anti-PD-1 [6–10 ], and anti-CTLA-4 [11 , 12 ], is increasingly appreciated. Although multiple studies have reported that a favorable gut microbiome is associated with response to anti-PD-1 in cancer patients, its precise composition is not yet fully understood [13–15 ], and the concordance among identified species in different studies is limited [13–17 ].
Given the role of gut microbiota in mediating nonresponse in ICI-treated melanoma, several clinical trials have studied the role of microbiome interventions in treating PD-1 nonresponsive cancers with interventions ranging from probiotics, single strain bacterial isolates (MRx0518 and EDP1503), defined bacterial consortia (VE800 and SER301) and FMT [18 ]. Of these, given the as-yet-undefined significant complexity underlying immune-tumor-microbiome interactions, FMT represents the most direct method of microbiome modulation by which beneficial bacteria can be introduced into an established complex ecosystem along with other microorganisms that may play supportive roles.
To evaluate the role of microbiome modulation in reversing primary resistance to anti-PD-1 ICI, we previously conducted a feasibility trial to evaluate the safety and efficacy of responder-derived FMT (R-FMT) together with anti-PD-1 pembrolizumab in PD-1 refractory metastatic melanoma patients [19 ]. In this study, we demonstrated that single administration of R-FMT resensitized patients with primary refractory melanoma to pembrolizumab: with objective responses in three patients and durable stable disease (SD) lasting >12 months in three patients (i.e. objective response rate 20% and disease control rate 40%) with a low rate of toxicity [19 ]. Response to R-FMT was associated with enrichment of taxa belonging to phyla Firmicutes and Actinobacteria in post-FMT fecal samples in recipients, along with reduced CXCL8/IL-8 expressing myeloid cells intratumorally, and increased CD56+CD8+ T cells, CD45RA/CD4 positive effector memory T cells and mucosal-associated invariant T cells peripherally [19 ]. Similar results were observed in another trial of R-FMT in PD-1 refractory melanoma [20 ].
These results have spurred the efforts to create local stool banks containing ready-to-use, high-quality, donor fecal material for use in immuno-oncologic studies such as the ones described above. Given concerns regarding the potential transmission of pathogenic organisms and other noninfectious traits putatively mediated by gut microbiome (metabolic syndrome and fibromyalgia, etc.); candidate stool donors and donor fecal material typically undergo extensive pretransplant screening and testing [21 ]. In the rCDI context, a recent study that evaluated the stool of candidate FMT donors to determine fitness for clinical use reported that only 3% of donors passed the quality control assessment [22 ]. Herein, we report on our experience in establishing a novel donor-screening program to generate R-FMT for use in PD-1 refractory melanoma. We report on the success rate of history and serologic screening, and seroprevalence of latent viral, bacterial and parasitic organisms in this unique cohort. We compare the donor-screening methodology used herein with other programs that have published screening methods and acceptance rates albeit in nononcology indications [23–27 ]. Finally, we review the cost and feasibility of establishing such a screening program.
Methods
PD-1 responder donor recruitment
Candidate PD-1 responder donors were recruited through clinic-based encounters at the Hillman Cancer Center in Pittsburgh, Pennsylvania which is affiliated with the University of Pittsburgh Medical Center (UPMC) hospital system, including UPMC Shadyside and UPMC Presbyterian Hospitals. Recruitment of donors was divided into two phases: pre-SARS-coronavirus-2 (SARS-CoV02) infection (COVID) and post-COVID. The pre-COVID phase of recruitment took place from May 2018 to December 2019; the post-COVID phase started in March 2021 and concluded in July 2022. Candidate PD-1 responder donors were identified based on a documented history of metastatic cutaneous (or unknown primary) melanoma treated with nivolumab, pembrolizumab or other approved/investigational anti-PD-(L)1 ICI administered singly. Enrollment was restricted to PD-1 responder patients who were currently in durable remission, defined as median duration of remission lasting ≥12 months (for complete responders) or ≥24 months (for partial responders) measured since initiation of therapy, as published data indicates that the likelihood of relapse was negligible in ICI-treated melanoma patients with the durable response at this duration of follow-up [28–30 ]. Other eligibility criteria included: absence of recent antibiotic treatment (defined as oral/parenteral within 1 month of donation); any history of significant gastrointestinal illnesses, including active inflammatory bowel disease, active irritable bowel syndrome, any chronic diarrheal disorder, active primary gastrointestinal malignancy or major gastrointestinal surgical procedures; history of symptomatic autoimmune illness; history of documented chronic pain syndromes (fibromyalgia or chronic fatigue); history of severe (35–39.9 kg/m2 ) or morbid (>40.0 kg/m2 ) obesity based on BMI; and active pregnancy. Patients with suspected SARS-CoV-2/COVID infection diagnosed per Center for Disease Control and Prevention (CDC) guidelines were excluded in the post-COVID phase of the study.
Donor health was continuously monitored throughout their donation lifetime. FMT studies in non-oncological indications have typically utilized bookend testing, wherein blood and stool tests are performed twice on either end of a collection period that typically spans 60 days. However, given the novel nature of this study (PD-1 refractory melanoma), the use of a previously uncharacterized source of FMT (PD-1 durable responders) and the uncertain performance of bookend testing in these circumstances, all tests were repeated every 14 days during the donor testing phase as illustrated in Fig. 1 Â (Fig. 1: Donor and Recipient Screening Protocol). Samples were quarantined until the donor passed screening at the end of the collection window.
Fig. 1: Donor and recipient screening protocol.
PD-1 responder donor screening
Candidate PD-1 responder donors who expressed a willingness to participate were screened for exposures to infectious agents using a previously published methodology for the use of FMT to treat rCDI,31 that consisted of a specific questionnaire (Supplementary Table 1, Supplemental digital content 1, https://links.lww.com/MR/A307 ). Candidate donors who declined participation were asked no further questions. If candidate PD-1 responder donors passed initial screening criteria, they were asked to provide written informed consent, received a full history and physical examination by a physician and underwent stool, blood and nasopharyngeal testing for potentially transmissible infectious agents serologic and fecal testing of infectious agents listed in Table 1 .
Table 1 -
Infectious screening assays on blood, stool and nasopharyngeal specimens
Infectious agent
Assay
Nature of sample (blood, stool other)
Lab processing test
Assay manufacturer
Interpretation of result
Cytomegalovirus (CMV)
CMV IgG
Blood
PUH ID Lab
Bio-Rad immunoassay (ToRc IgG)
Seromatching required between donors/recipients.
Epstein-Barr virus (EBV)
EBV IgG and IgM
Blood
PUH ID Lab
Bio-Rad immunoassay (EBV IgG and IgM)
Seromatching required between donors/recipients.
Entameba histolytica
ELISA
Blood
Quest Diagnostics via SHY ID Lab
Quest
•  Excluded from donating if positive.
•  Referral to infectious diseases if infected.
Hepatitis A virus (HAV)
HAV total antibody (anti-HAV IgM and IgG)
Blood
Quest diagnostics via SHY ID Lab
Quest immunoassay
•  Excluded from donating if positive.
•  Referral to infectious diseases.
Hepatitis B virus (HBV)
HBV surface antigen (HBsAg), HBV surface antibody (anti-HBs), and HBV core antibody (anti-HBc)
Blood
PUH ID Lab
Siemens immunoassay (HBsAg, HBc Total, anti-HBs2)
•  Only immune and non-immune/uninfected individuals permitted to donate.
•  Referral to infectious diseases if infected.
Hepatitis C virus (HCV)
Hepatitis C antibody (anti-HCV) with reflex HCV RNA
Blood
PUH ID Lab
Siemens immunoassay (HCV) with reflex confirmation using Quest
•  Excluded from donating if positive (anti-HCV reactive, and HCV RNA positive)
•  Referral to infectious diseases
Herpes simplex virus (HSV) type 1 (HSV-1)
HSV-1 IgG
Blood
PUH ID Lab
Bio-Rad immunoassay (HSV-1/2 IgG)
Seromatching required between donors/recipients.
Herpes simplex virus (HSV) type 2 (HSV-2)
HSV-2 IgG
Blood
PUH ID Lab
Bio-Rad immunoassay (HSV-1/2 IgG)
Seromatching required between donors/recipients.
Human herpesvirus 6 (HHV-6)
HHV-6 IgG
Blood
Quest diagnostics via SHY ID Lab
Quest immuno-fluorescence assay
Seromatching required between donors/recipients.
Human polyomavirus-2 (JC virus)
JC virus RT PCR
Blood
Quest diagnostics via SHY ID Lab
Quest RT-PCR
Seromatching required between donors/recipients.
Human T-lymphotropic virus (HTLV)-1
HTLV-1 IgG
Blood
Quest diagnostics via SHY ID Lab
Screen immunoassay, confirmatory immunoblot Quest
•  Excluded from donating if positive.
•  Referral to infectious diseases.
Human T-lymphotropic virus (HTLV)-2
HTLV-2 IgG
Blood
Quest diagnostics via SHY ID Lab
Screen immunoassay, confirmatory immunoblot Quest
•  Excluded from donating if positive.
•  Referral to infectious diseases.
Human immunodeficiency virus (HIV)
5th generation IgG with reflex ELISA
Blood
PUH ID Lab
Bio-Rad immunoassay (HIV Ag-Ab)
•  Excluded from donating if positive.
•  Referral to infectious diseases.
Strongyloides stercoralis
S. stercoralis IgG
Blood
Quest diagnostics via SHY ID Lab
Quest immunoassay
•  Excluded from donating if positive.
•  Referral to Infectious Diseases.
Syphilis
RPR, IgM, IgG
Blood
PUH ID Lab
T pallidum antibody immunoassay reflex RPR screen Bio-Rad
•  Excluded from donating if positive.
•  Referral to infectious diseases.
Campylobacter species
Culture
Stool
PUH ID Lab
Diasorin, Verigene
•  Excluded from donating if positive.
Clostridium difficile (C. diff)
C. diff toxin EIA with reflex PCR
Stool
PUH ID Lab
EIA (Alere), PCR (Cepheid)
•  Excluded from donating if positive.
Helicobacter pylori (H. pylori)
EIA
Stool
Quest diagnostics via SHY ID Lab
Quest
•  Excluded from donating if positive.
Salmonella
Culture
Stool
PUH ID Lab
Diasorin, Verigene
•  Excluded from donating if positive.
Shigella
Culture
Stool
PUH ID Lab
Diasorin, Verigene
•  Excluded from donating if positive.
Vibrio
Culture
Stool
PUH ID Lab
Diasorin, Verigene
•  Excluded from donating if positive.
Yersinia
Culture
Stool
PUH ID Lab
Diasorin, Verigene
•  Excluded from donating if positive.
Rotavirus
Culture
Stool
PUH ID Lab
Diasorin, Verigene
•  Excluded from donating if positive.
•  May be reconsidered for donation after 12 weeks from the initial positive test, if the subsequent test is negative.
Adenovirus
Adenovirus DNA RT PCR
Stool
Quest diagnostics via SHY ID Lab
Quest
•  Excluded from donating if positive.
•  May be reconsidered for donation after 12 weeks from the initial positive test, if the subsequent test is negative.
Norovirus
Culture
Stool
PUH ID Lab
Diasorin, Verigene
•  Excluded from donating if positive.
•  May be reconsidered for donation after 12 weeks from the initial positive test, if the subsequent test is negative.
Giardia
EIA
Stool
PUH ID Lab
Quik Chek, Alere
•  Excluded from donating if positive.
•  May be reconsidered for donation after 12 weeks from the initial positive test, if the subsequent test is negative.
Cryptosporidia
EIA
Stool
PUH ID Lab
Quik Chek, Alere
•  Excluded from donating if positive.
•  May be reconsidered for donation after 12 weeks from the initial positive test, if the subsequent test is negative.
Enterovirus
RT-PCR
Stool
Quest diagnostics via SHY ID Lab
Quest
•  Excluded from donating if positive.
Enterohemorrhagic E. Coli (EHEC)
NAAT
Stool
Quest Diagnostics via SHY ID Lab
Quest
•  Excluded from donating if positive.
Vancomycin-resistant Enterococcus (VRE)
Culture
Stool
PUH ID Lab
SpectraVRE chromagar, Remel
•  Excluded from donating if positive.
Carbapenem-resistant Enterobacteriaceae (CRE)
Culture
Stool
PUH ID Lab
CRE VRE chromagar, Hardy Diagnostics
•  Excluded from donating if positive.
Extended-spectrum β-lactamase-producing Enterobacteriaceae (ESBL)
Culture
Stool
PUH ID Lab
ESBL chromagar, Hardy Diagnostics
•  Excluded from donating if positive.
SARS-CoV-2
COVID-19 rtPCR
Stool
DSL Laboratories
DSL Laboratories
•  Excluded from donating if positive
•  Prohibited from donating for 12 weeks following this exposure.
•  Before being reconsidered for donations, patients must undergo bookend testing as outlined and test negative.
SARS-CoV-2
COVID-19 rtPCR
Nasopharyngeal swab
DSL Laboratories
DSL Laboratories
•  Excluded from donating if positive
•  Prohibited from donating for 12 weeks following this exposure.
•  Before being reconsidered for donations, patients must undergo bookend testing as outlined and test negative.
Methicillin-resistant Staphylococcus aureus (MRSA)
MRSA culture
Nasopharyngeal swab
Quest Diagnostics via SHY ID Lab
MRSA chromagar
•  Excluded from donating if positive.
Presbyterian Infectious Disease (PUH ID); Shadyside Infectious Disease (SHY ID)
Potential donors were screened for the following transmissible agents in blood: fourth-generation combination HIV antigen and antibody immunoassay, human T-cell lymphotropic virus (HTLV) type 1 and 2, hepatitis A, hepatitis B, hepatitis C, cytomegalovirus, Epstein-Barr virus (EBV), syphilis, herpes simplex virus (HSV) type 1 and 2, human herpesvirus-6 (HHV-6), human polyomavirus-2 (JC virus), Strongyloides stercoralis and Entamoeba histolytica . The stool was screened for Campylobacter, Escherichia coli H7, Salmonella, Shigella, Vibrio, Yersinia, Norovirus, Rotavirus, Clostridium difficile toxin (with reflex PCR), Helicobacter pylori, Adenovirus, Giardia, Cryptosporidia , vancomycin-resistant enterococci (VRE), carbapenem-resistant enterobacteriacea (CRE) and extended-spectrum beta-lactamase (ESBL) bacterial species. PD-1 responder donors were additionally tested for nasal methicillin-resistant Staphylococcus aureus (MRSA). Pregnancy tests were conducted if the patient was female and not postmenopausal.
PD-1 responder donor stool sample processing and storage
PD-1 responder donor stool specimens were obtained every 14 days as illustrated in Fig. 1  (Fig. 1: Donor and Recipient Screening Protocol). A portion of each labeled specimen was used for infectious testing as outlined above and all stool specimens were processed in an anerobic hood. To create one FMT infusate, approximately 30g of donor stool was weighed on a calibrated digital scale. Sufficient sterile physiological saline (0.9% w/v of NaCl) and 85% glycerol were added for a final concentration of 10% of the fecal suspension. Fecal suspension was resuspended using a sterile spatula, passed through a 100μm cell strainer to remove particulate matter. The recovered fecal infusate was stored in a designated freezer at -800 C for up to 6 months. Prior to use, frozen fecal infusates were thawed for 20-30 minutes in a 25°C waterbath and transferred in a 50ml syringe with catheter tip. Unused samples beyond 6 months were discarded. Chain of custody logs that included details of all individuals who received and relinquished samples from processing till administration were maintained.
Results
Performance of screening algorithm and SARS-CoV-2 modification
Donor health was continuously monitored throughout their donation lifetime. FMT studies in nononcological indications have typically utilized bookend testing, wherein blood and stool tests are performed twice on either end of a collection period that typically spans 60 days. However, given the novel nature of this study (PD-1 refractory melanoma), the use of a previously uncharacterized source of FMT (PD-1 durable responders) and the uncertain performance of bookend testing in these circumstances, all tests were repeated every 14 days during the donor testing phase as illustrated in Fig. 1 . Samples were quarantined until the donor passed screening at the end of the collection window.
Donors who tested positive for serious blood-borne pathogens, enteric pathogens and multi-drug resistant organisms were permanently excluded from donation as illustrated in Fig. 2 . Donors who screened positive for enteric pathogens (Rotavirus, Adenovirus, Norovirus, Giardia, Cryptosporidium and Enterovirus ) were excluded regardless of symptoms but could be reconsidered following resolution of acute infection if they were asymptomatic, resumed normal stool patterns and repeat testing procedures were negative. Donors and recipients were sero-matched for latent viruses (cytomegalovirus, EBV, Hepatitis A/B/C, herpes simplex type 1/2, human herpesvirus 6, JC virus) as previously described [31 ]. Eligible PD-1 R donors provided repeat donations and underwent retesting every 2 weeks to ensure adequate sample availability in the stool bank (Fig. 1 ).
Fig. 2: Donor and recipient matching protocol.
Following the outbreak of COVID-19, this clinical trial was initially halted before the development of a COVID-19/SARS-CoV-2 testing algorithm in discussion with the FDA and based on consensus guidelines [32–34 ] (Fig. 1 ). Given the uncertainties over potential SARS-CoV-2 fecal-oral transmission, the prior testing algorithm was altered to include additional screening questions to ascertain SARS-CoV-2 exposure in the donor/recipient questionnaire (questions 39–41, Supplementary Table 1, Supplemental digital content 1, https://links.lww.com/MR/A307 ), and patients were clinically assessed for COVID-19 by querying for fever, cough, dyspnea, chills, anosmia or ageusia, sore throat, muscle pain not otherwise explainable by alternative diagnosis within the previous 30 days per CDC recommendations [35 ]. Additionally, stool and nasopharyngeal specimens from both donors and recipients were tested using an FDA Emergency Use Authorization (EUA)-approved, Clinical Laboratory Improvement Amendments-certified COVID-19 diagnostic assay from the Diagnostic Solutions Laboratory. This reverse transcription PCR (RT-PCR) assay permitted qualitative detection of SARS-CoV-2 nucleic acids. Determination of ‘SARS-CoV-2 negative’ status was made on nasopharyngeal and stool specimens obtained at two timepoints: 1st sample was collected at least 14 days before stool sample donation, and the 2nd sample was collected at least 14 days after stool sample donation. The stool sample was released for FMT use no sooner than 48–72 h after 2nd test resulted and donors (and recipients) were deemed ‘SARS-CoV-2 negative’ (Fig. 1 ). Samples with discordant stool and nasopharyngeal SARS-CoV-2 results were excluded. Candidate donors (and recipients) who tested ‘SARS-CoV-2 positive’ were referred to infectious disease physicians for further treatment.
Twenty-nine candidate PD-1 responder donors were evaluated, all of Caucasian ethnicity. Of these 29 donors, 26 passed prescreening and had a serological evaluation of blood and stool as summarized in Fig. 3 . The three patients excluded in prescreening had BMI >35. Following initial serological studies, nine patients were excluded following the identification of potentially transmissible serious infections including hepatitis A (n  = 6), hepatitis B, hepatitis C and syphilis (1 each) (Fig. 3 ). Each of these candidate donors was subsequently referred to infectious disease specialists for dedicated treatment and were successfully treated but not considered eligible as donors. Three patients with documented E. histolytica were successfully treated with antiparasitic agents but were also excluded as donors. Candidate PD-1 R donors who met all criteria and screened negative for serious infections including COVID-19/SARS-CoV-2 (in post-COVID period) were designated ‘PD-1 responder eligible donors’. Therefore, after the full screening of 29 candidate PD-1 responder donors, 14 PD-1 responder donors were identified as donors eligible to donate on a regular basis for the program for an acceptance rate of 48% (14/29).
Fig. 3: Consort Diagram Depicting Candidate Donors During Protocol.
Results of infectious serologies on stool, blood and nasopharyngeal samples from donors and recipients
Among PD-1 responder donors, we observed the following seropositivity rates at pre- and post-COVID timepoints, respectively: cytomegalovirus (35.7 and 41.2%), EBV (96.4 and 81.5%), HSV-1 (32.1 and 37.0%), HSV-2 (14.3 and 3.7%), HHV-6 (100.0 and 92.6%) and JC virus (85.7 and 96.3%) (Table 2 ). Given that cancer patients are typically screened for exposure to chronic infectious hepatitis and HIV before exposure to anti-PD-1, seropositivity rates for Hepatitis B, C and HIV were unsurprisingly 0.0% at both pre- and post-COVID timepoints, respectively in both donors and recipients (Table 2 ). These results are similar to published seroprevalence statistics from healthy human blood donors in the USA – cytomegalovirus (45–100%) [36–38 ], EBV (80–100%) [39 , 40 ], HSV-2 (3–17%) [38 , 41–43 ] and HHV-6 (98–100%) [38 , 44 ]. However, the seroprevalence of HSV-1 in our cohort (32–37%) was lower than reported (67–82%) [38 , 41–43 ]. Conversely, the seroprevalence of the JC virus in our cohort (86–96%) was higher than reported (57–86%) [45 , 46 ]. Concordant with published seroprevalence statistics [47–49 ], seropositivity rates for HTLV-1, HTLV-2 and syphilis were very low at both pre- and post-COVID timepoints, respectively (Table 2 ).
Table 2 -
Results of infectious screening assays (blood)
Infectious Agent
Assay
Pre-COVID Donors
Pre-COVID Recipients
Post-COVID Donors
Post-COVID Recipients
Seropositive rate
Seronegative rate
n
Seropositive rate
Seronegative rate
n
Seropositive rate
Seronegative rate
n
Seropositive rate
Seronegative rate
n
Blood
 Cytomegalovirus (CMV)
CMV IgG
35.7%
64.3%
28
41.2%
58.8%
17
3.7%
96.3%
27
0.0%
100.0%
1
 Epstein Barr virus (EBV)
EBV IgG and IgM
96.4%
3.6%
28
100.0%
0.0%
17
81.5%
18.5%
27
100.0%
0.0%
1
 Entameba histolytica
E. histolytica IgG
3.6%
96.4%
28
0.0%
100.0%
17
22.2%
77.8%
27
0.0%
100.0%
1
 Hepatitis A virus (HAV)
HAV total antibody (anti-HAV IgM and IgG)
0.0%
100.0%
28
23.5%
76.5%
17
11.1%
88.9%
27
100.0%
0.0%
1
 Hepatitis B virus (HBV)
HBV surface antibody
3.6%
96.4%
28
11.8%
88.2%
17
0.0%
100.0%
27
0.0%
100.0%
1
 Hepatitis B virus (HBV)
HBV surface antigen
0.0%
100.0%
28
0.0%
100%
17
0.0%
100.0%
27
0.0%
100.0%
1
 Hepatitis B virus (HBV)
HBV core antibody
0.0%
100.0%
28
5.9%
94.1%
17
0.0%
100.0%
27
0.0%
100.0%
1
 Hepatitis C virus (HCV)
Hepatitis C antibody
0.0%
100.0%
28
0.0%
100.0%
17
0.0%
100.0%
27
0.0%
100.0%
1
 Herpes simplex virus (HSV) type 1 (HSV-1)
HSV-1 IgG
32.1%
67.9%
28
64.7%
35.3%
17
37.0%
63.0%
27
100.0%
0.0%
1
 Herpes simplex virus (HSV) type 2 (HSV-2)
HSV-2 IgG
14.3%
85.7%
28
29.4%
70.6%
17
3.7%
96.3%
27
0.0%
100.0%
1
 Human herpesvirus 6 (HHV-6)
HHV-6 IgG
100.0%
0.0%
28
76.5%
23.5%
17
92.6%
7.4%
27
100.0%
0.0%
1
 Human polyomavirus 2 (JC virus)
JC virus IgG
85.7%
14.3%
28
88.2%
11.8%
17
96.3%
3.7%
27
100.0%
0.0%
1
 Human T-lymphotropic virus (HTLV)-1
HTLV-1 IgG
0.0%
100.0%
28
0.0%
100.0%
17
0.0%
100.0%
27
0.0%
100.0%
1
 Human T-lymphotropic virus (HTLV)-2
HTLV-2 IgG
0.0%
100.0%
28
0%
100.0%
17
0.0%
100.0%
27
0.0%
100.0%
1
 HIV
5th generation IgG with reflex ELISA
0.0%
100.0%
28
0%
100.0%
17
0.0%
100.0%
27
0.0%
100.0%
1
 Strongyloides stercoralis
S. stercoralis IgG
0.0%
100.0%
28
0%
100.0%
17
0.0%
100.0%
27
0.0%
100.0%
1
 Syphilis
RPR, IgM, IgG
0.0%
100.0%
28
0%
100.0%
17
0.0%
100.0%
27
0.0%
100.0%
1
We extensively evaluated donors and recipients for multiple enteric pathogens including S. stercoralis and E. histolytica in blood; and Campylobacter, E. coli O157:H7, Salmonella, Shigella, Vibrio, Yersinia, Norovirus, Rotavirus, C. difficile, H. pylori, adenovirus, Giardia and Cryptosporidia. We noted a 3.6% incidence of E. histolytica , 3.6% incidence of norovirus, 3.7% incidence of giardia and 7.1% incidence of C. difficile in donors across pre- and post-COVID timepoints (Tables 2 and 3 ). These particular samples were discarded, the patients referred for specific therapy where appropriate, and upon re-screening, the patients were found to be negative although these donors were excluded. No recipients tested positive for any enteric pathogens. We also evaluated both donors and recipients for MDROs (CRE, VRE, ESBL) in stool and MRSA in nasopharyngeal samples. The incidence of MDROs including MRSA among donors and recipients was 0.0%, lower than reported point prevalence rates of MRSA colonization among hospital-exposed patients in an urban, nonoutbreak setting [50 , 51 ].
Table 3 -
Results of infectious screening assays (stool)
Infectious agent
Assay
Pre-COVID Donors
Pre-COVID Recipients
Post-COVID Donors
Post-COVID Recipients
Seropositive rate
Seronegative rate
n
Seropositive rate
Seronegative rate
n
Seropositive rate
Seronegative rate
n
Seropositive rate
Seronegative rate
n
Stool
 Campylobacter species
GI pathogen panel
0%
100%
28
0%
100%
17
0%
100%
27
0%
100%
1
 Clostridium difficile (C. diff)
C. diff toxin with reflex PCR
7.1%
92.9%
28
0%
100%
17
0%
100%
27
0%
100%
1
 Escherichia coli O157:H7
E. coli O157:H7
0%
100%
28
0%
100%
17
0%
100%
27
0%
100%
1
 Helicobacter pylori (H. pylori)
H. pylori antigen with reflex ELISA
0%
100%
28
0%
100%
17
0%
100%
27
0%
100%
1
 Salmonella
GI pathogen panel
0%
100%
28
0%
100%
17
0%
100%
27
0%
100%
1
 Shigella
GI pathogen panel
0%
100%
28
0%
100%
17
0%
100%
27
0%
100%
1
 Vibrio
GI pathogen panel
0%
100%
28
0%
100%
17
0%
100%
27
0%
100%
1
 Yersinia
GI pathogen panel
0%
100%
28
0%
100%
17
0%
100%
27
0%
100%
1
vRotavirus
GI pathogen panel
0%
100%
28
5.9%
94.1%
17
0%
100%
27
0%
100%
1
 Adenovirus
Adenovirus DNA PCR
0%
100%
28
0%
100%
17
0%
100%
27
0%
100%
1
 Norovirus
GI pathogen panel
3.6%
96.4%
28
0%
100%
17
0%
100%
27
0%
100%
1
 Giardia
Ova and parasite screen
0%
100%
28
0%
100%
17
3.7%
96.3%
27
0%
100%
1
 Cryptosporidia
Ova and parasite screen
0%
100%
28
0%
100%
17
0%
100%
27
0%
100%
1
 Vancomycin-resistant Enterococcus (VRE)
VRE screen
0%
100%
28
0%
100%
17
0%
100%
27
0%
100%
1
 Carbapenem-resistant Enterobacteriaceae (CRE)
CRE screen
0%
100%
28
5.9%
94.1%
17
0%
100%
27
0%
100%
1
 Extended-spectrum β-lactamase-producing Enterobacteriaceae (ESBL)
ESBL screen
0%
100%
28
5.9%
94.1%
17
0%
100%
27
0%
100%
1
 SARS-CoV-2
COVID-19 rtPCR
N/A
N/A
N/A
N/A
N/A
N/A
11.1%
88.9%
27
0%
100%
1
Following the COVID-19 epidemic and reports of excretion and viability of SARS-CoV-2 in feces and association with infection [52 , 53 ], we modified the donor/recipient questionnaire to include additional screening questions to evaluate for SARS-CoV-2 exposure (questions 39–41, Supplementary Table 1, Supplemental digital content 1, https://links.lww.com/MR/A307 ), and additionally screened for SARS-CoV-2 both by nasopharyngeal swab and in stool using an FDA EUA-approved RT-PCR assay. The incidence of SARS-CoV-2 in stool was 11.1% in donors (3 of 27 specimens) and 0.0% in recipients; and the incidence in nasopharyngeal samples was 0.0% in donors and 0.0% in recipients (Tables 3 and 4 ). Of note, the three donors who tested positive for SARS-CoV-2 did so only on the stool and not nasopharyngeal swabs. All three donors had screened negative on the screening questionnaire, were afebrile, otherwise asymptomatic and had no SARS-CoV-2-specific recent exposures. Per CDC guidance, these samples were discarded and patients were advised to self-quarantine. All three donors were rescreened 12 weeks later and were negative for SARS-CoV-2 in both stool and nasopharyngeal specimens. The 11.1% positivity rate among candidate stool donors for an FMT program has not previously been reported.
Table 4 -
Results of infectious screening assays (nasopharynx)
Infectious agent
Assay
Pre-COVID Donors
Pre-COVID Recipients
Post-COVID Donors
Post-COVID Recipients
Seropositive rate
Seronegative rate
n
Seropositive rate
Seronegative rate
n
Seropositive rate
Seronegative rate
n
Seropositive rate
Seronegative rate
n
Nasal
 MRSA
0%
100%
28
0%
100%
17
0%
100%
27
0%
100%
1
 SARS-CoV-2
COVID-19 RT-PCR
N/A
N/A
N/A
N/A
N/A
N/A
0%
100%
27
0%
100%
1
Cost of infectious testing
The cost of a full infectious work-up at our center was approximately US $2260.24 per donor per timepoint not including SARS-CoV-2 testing, and US $2460.24 per donor per timepoint if SARS-CoV-2 testing was included (Table 5 ). The total costs of screening all donors and 16 recipients were $129 713.20 and $40 884.32, respectively; and the cost of identifying a single eligible donor was $9 265.23.
Table 5 -
Cost of donor and/or recipient screening
Infectious agent
Assay
CPT codes
Cost (per patient per time point)
Blood
 Cytomegalovirus (CMV)
CMV IgG
86644
25.00
 Epstein Barr virus (EBV)
EBV IgG and IgM
866650000, 866650000
27.86
 Entameba histolytica
E. histolytica IgG
867530000
143.00
 Hepatitis A virus (HAV)
HAV total antibody (anti-HAV IgM and IgG)
86708 & 86709
42.00
 Hepatitis B virus (HBV)
HBV surface antibody
86706
18.00
 Hepatitis B virus (HBV)
HBV surface antigen
87340
18.00
 Hepatitis B virus (HBV)
HBV core antibody
86704
21.00
 Hepatitis C virus (HCV)
Hepatitis C antibody
86803
25.00
 Herpes simplex virus (HSV) type 1 (HSV-1)
HSV-1 IgG
86694 and 86695
31.95
 Herpes simplex virus (HSV) type 2 (HSV-2)
HSV-2 IgG
86696
34.00
 Human herpesvirus 6 (HHV-6)
HHV-6 IgG
875320000
511.00
 Human polyomavirus 2 (JC virus)
JC virus IgG
877980000
46.00
 Human T-lymphotropic virus (HTLV)-1
HTLV-1 IgG
86687
15.00
 Human T-lymphotropic virus (HTLV)-2
HTLV-2 IgG
86688
25.00
 Human immunodeficiency virus (HIV)
5th generation IgG with reflex ELISA
86703 & 87390
55.00
 Strongyloides stercoralis
S. stercoralis IgG
866820000
10.67
 Syphilis
RPR, IgM, IgG
867800000
8.69
Stool
 Campylobacter species
GI pathogen panel
870460000
2.34
 Clostridium difficile (C. diff)
C. diff toxin with reflex PCR
87324
20.00
 Escherichia coli O157:H7
E. coli O157:H7
870460000
1.48
 Helicobacter pylori (H. pylori)
H. pylori antigen with reflex ELISA
87338
24.00
 Salmonella
GI pathogen panel
870450000
5.23
 Shigella
GI pathogen panel
870450000
5.23
 Vibrio
GI pathogen panel
87046
2.34
 Yersinia
GI pathogen panel
870460000
1.40
 Rotavirus
GI pathogen panel
86759
23.00
 Adenovirus
Adenovirus DNA PCR
877980000
350.00
 Norovirus
GI pathogen panel
877990000
736.00
 Giardia
Ova and parasite screen
87329
21.00
 Cryptosporidia
Ova and parasite screen
87328
21.00
 Vancomycin-resistant Enterococcus (VRE)
VRE screen
870810000
6.00 (bundled with CRE, ESBL)
 Carbapenem-resistant Enterobacteriaceae (CRE)
CRE screen
870810000
 Extended-spectrum β-lactamase-producing Enterobacteriaceae (ESBL)
ESBL screen
870810000
 SARS-CoV-2
SARS-CoV-2
86769
100.00
Nasopharynx
 Methicillin-resistant Staphylococcus aureus
MRSA screening culture
87641
62.00
 SARS-CoV-2
SARS-CoV-2
86769
100.00
Total Cost (Pre-COVID, all donors)
63 286.72
Total Cost (Post-COVID, all donors)
66 426.48
Total Cost (Pre-COVID, all recipients)
38 424.08
Total Cost (Post-COVID, all recipients)
2460.24
Total Cost (Pre-COVID, per donor/recipient)
2260.24
Total Cost (Post-COVID, per donor/recipient)
2460.24
Discussion
The link between gut dysbiosis and both immune-related adverse events and nonresponse to cancer immunotherapy across multiple settings including anti-PD-1 ICI and anti-CD19 chimeric antigen receptor T-cell therapy is increasingly appreciated [10 , 54 , 55 ]. This has spurred multiple clinical trials aimed at evaluating various microbiome therapeutics including healthy donor or responder-derived FMT; complete consortia products (SER-401, NCT03817125; MaaT033, NCT03772899); synthetic bacterial consortia (VE800, NCT04208958) and monoclonal microbials (CBM588, NCT03829111 and NCT05122546; EDP1503, NCT03595683; MRx0518, NCT03775850). Given the early data demonstrating the efficacy of responder FMT in treating PD-1 nonresponsive melanoma, there is great interest in studying this in other oncologic indications and multiple FMT trials are ongoing in various settings including mismatch repair deficient (dMMR) tumors following progression on anti-PD-1 ICI (NCT04729322), gastrointestinal cancers following progression on anti-PD-1 ICI (NCT04130763) and treatment-naĂ¯ve metastatic melanoma patients in combination with anti-PD-1 ICI (NCT03772899). Hence, there is an increasing demand to establish and maintain FMT donor pools at individual cancer centers.
Of the initial 29 donors who were screened, following the exclusion of three for excessive BMI, and 12 for infectious issues, 14 eligible donors were identified, eight of whom were utilized in the clinical trial. The overall efficiency of screening was 48% (14/29) and higher than what has been reported with FMT programs to treat refractory C. difficile infection at single centers (10–37%) [23 , 24 , 26 ], and considerably higher than the <3% reported by OpenBiome, the longest standing international stool bank [56 ]. The observed screening efficiency, which was not significantly impacted by the COVID-19 pandemic, suggests that a well-resourced, organized, screening program based out of an academic institution should be able to generate sufficient responder-derived donor material for clinical trial purposes.
In the context of healthy donor FMT programs established to treat rCDI, the cost of screening 1 donor has been reported as ranging from US $440 to US $885 [24 , 25 ]. Herein, we report that the cost of screening a single donor was US $2460.24 per donor per timepoint or $9265.23 to identify a single eligible donor. The greater costs reported herein are likely related to the extensive infectious serological studies (Table 1 ). Overall, this argues that the cost of developing a responder-derived donor fecal bank is considerable and far greater than similar approaches using healthy donor material, particularly if commercially sourced from stool banks such as OpenBiome.
In hematological malignancy patients following allogeneic hematopoietic cell transplant (allo-HCT), FMT post-allo-HCT is associated with the expansion of donor-derived taxa and restoration of gut microbiota diversity [57 , 58 ]. Separately, FMT has been successfully investigated as a treatment for steroid-refractory acute GI graft-versus-host disease as well [59 , 60 ]. However, even in this immunocompromised population with paramount concerns for infectious complications, the incidence of FMT-related bacteremia is low [57 ]. Separately, no instances of FMT-related infectious complications were reported in either of the two immuno-oncologic studies that evaluated FMT administration to advanced cancer patients along with anti-PD-1 in refractory melanoma [19 , 20 ]. Given the low observed prevalence of certain GI pathogens (such as Campylobacter, H. pylori, Salmonella, Shigella, Yersinia, Adenovirus, Cryptosporidia , etc.), it may be preferable to use a bookend testing procedure (i.e. testing before and after multiple stool donations) with an appropriate window (i.e. 60 days) while quarantining stool samples until the post-donation tests are confirmed negative.
We observed an 11.1% positivity rate of asymptomatic SARS-CoV-2 in the stool of candidate FMT donors. Given that SARS-CoV-2 can be found in feces [61 ], the potential risk of fecal transmissibility [62 ], and that these patients denied significant exposures and screened negative for concerning symptoms, the reported positivity rate strongly argues for repeated screening of donor stools through a molecular test for SARS-CoV-2 on both stool and nasopharyngeal specimens at each donation period with a relatively short window such as 14 days. It is important for the FMT stool banks, FMT clinical team, treating physicians and FMT recipients to recognize that a negative test for a pathogen does not fully mitigate the risk of transmission via donated material. Concurrently, it is equally important for FMT stool banks and FMT clinical teams to periodically update their laboratory testing as more sensitive and/orspecific tests are developed and as new infectious agents are deemed transmissible via the fecal route (e.g. monkeypox virus) [63 ].
Limitations of this study include the small sample size and the reasons for exclusion which may not necessarily be generalizable to other nononcology patient populations. The screening costs were accurate as this is a single-center study and the information pertaining to codes was readily available, but may not be applicable to non-US locations.
There is growing interest in developing microbiome-based therapeutic approaches in immuno-oncology to treat ICI resistance. Unlike the rCDI setting where the use of healthy donor FMT from central stool banks with storage and shipment of frozen samples is cost-effective and feasible [64 ], the data in the immuno-oncologic setting has been generated primarily with ICI responders and not healthy donors. Hence the development of an ICI responder-derived FMT program must consider the factors implicit in the care of a cancer patient including the need for longitudinal follow-up and the oncologist-patient relationship; suggesting that such programs may be better suited to being locally situated rather than centrally organized. These data suggest that the creation and maintenance of an ICI responder-derived FMT program are feasible. Full transparency and reporting of screening protocols are critical to enable clinicians and regulators to determine the acceptability and efficacy of such stool banks and further development of ICI responder-derived FMT programs.
Acknowledgements
The authors thank the patients and their caregivers who participated in this study, as well as the research teams at the UPMC Hillman Cancer Center. The research presented herein was funded in part through the Melanoma Research Foundation Breakthrough Consortium Award (D.D.), Jack Buncher Foundation (D.D.), NCI ETCTN Pittsburgh Cancer Consortium (PCC) Support Grant UM1 CA186690 (D.D.), NIH/NCI U01 CA268806 (H.M.Z. and D.D.) and R01 CA222203 (H.M.Z).
IRB approval number CR19060111-007, Hillman Cancer Center protocol number 17-034.
D.F.: data curation, formal analysis, writing–original draft. M.P.A.: data curation, formal analysis. D.H., M.S., H.D., C.H., S.B., S E., and A.R.: data curation. H.Z.: investigation, resources, data curation. D.D.: supervision, investigation, resources, data curation, formal analysis, writing–original draft, review and editing.
Conflicts of interest
D.D.: research support: Arcus, Bristol-Myers Squibb, Checkmate Pharmaceuticals, CellSight Technologies, Merck, GlaxoSmithKline/Tesaro, Zucero Therapeutics; Scientific Advisory Board: Vedanta Biosciences; consultancy with compensation: Checkmate Pharmaceuticals, Finch Inc., Immunocore, Shionogi and Vedanta Biosciences; Patents: (both provisional) U.S. Patent Application No. 63/208,719 (Compositions and Methods for Determining Responsiveness to Immune Checkpoint Inhibitors (ICI), Increasing Effectiveness of ICI, and Treating Cancer), and Application No. 63/124,231 (Compositions and Methods For Treating Cancer). H.Z.: Bristol-Myers Squibb, Checkmate Pharmaceuticals, and GlaxoSmithKline (research support) and Bristol-Myers Squibb, Checkmate Pharmaceuticals, GlaxoSmithKline, and Vedanta (consulting).
For the remaining authors, there are no conflicts of interest.
References
1. Chang JY, Antonopoulos DA, Kalra A, Tonelli A, Khalife WT, Schmidt TM, et al. Decreased diversity of the fecal microbiome in recurrent clostridium difficile-associated diarrhea. J Infect Dis 2008; 197:435–438.
2. Cammarota G, Masucci L, Ianiro G, BibbĂ² S, Dinoi G, Costamagna G, et al. Randomised clinical trial: faecal microbiota transplantation by colonoscopy vs. vancomycin for the treatment of recurrent Clostridium difficile infection. Aliment Pharmacol Ther 2015; 41:835–843.
3. Kelly CR, Khoruts A, Staley C, Sadowsky MJ, Abd M, Alani M, et al. Effect of fecal microbiota transplantation on recurrence in multiply recurrent clostridium difficile infection: a randomized trial. Ann Intern Med 2016; 165:609–616.
4. Ooijevaar RE, Van Nood E, Goorhuis A, Terveer EM, Van Prehn J, Verspaget HW, et al. Ten-year follow-up of patients treated with fecal microbiota transplantation for recurrent clostridioides difficile infection from a randomized controlled trial and review of the literature. Microorganisms 2021; 9:548.
5. van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, et al. Duodenal infusion of donor feces for recurrent clostridium difficile. N Engl J Med 2013; 368:407–415.
6. Dzutsev A, Goldszmid RS, Viaud S, Zitvogel L, Trinchieri G. The role of the microbiota in inflammation, carcinogenesis, and cancer therapy. Eur J Immunol 2015; 45:17–31.
7. Finlay BB, Goldszmid R, Honda K, Trinchieri G, Wargo J, Zitvogel L. Can we harness the microbiota to enhance the efficacy of cancer immunotherapy? Nat Rev Immunol 2020; 20:522–528.
8. Goldszmid RS, Dzutsev A, Viaud S, Zitvogel L, Restifo NP, Trinchieri G. Microbiota modulation of myeloid cells in cancer therapy. Cancer Immunol Res 2015; 3:103–109.
9. Zarour HM. Reversing T-cell dysfunction and exhaustion in cancer. Clin Cancer Res 2016; 22:1856–1864.
10. McCulloch JA, Davar D, Rodrigues RR, Badger JH, Fang JR, Cole AM, et al. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat Med 2022; 28:545–556.
11. Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol 2017; 28:1368–1379.
12. Vetizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015; 350:1079–1084.
13. Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018; 359:97–103.
14. Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre M-L, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018; 359:104–108.
15. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018; 359:91–97.
16. Frankel AE, Coughlin LA, Kim J, Froehlich TW, Xie Y, Frenkel EP, et al. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia 2017; 19:848–855.
17. Peters BA, Wilson M, Moran U, Pavlick A, Izsak A, Wechter T, et al. Relating the gut metagenome and metatranscriptome to immunotherapy responses in melanoma patients. Genome Med 2019; 11:61.
18. Matson V, Chervin CS, Gajewski TF. Cancer and the microbiome-influence of the commensal microbiota on cancer, immune responses, and immunotherapy. Gastroenterology 2021; 160:600–613.
19. Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin J-M, Morrison RM, et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science 2021; 371:595–602.
20. Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 2021; 371:602–609.
21. Carlson PE Jr. Regulatory considerations for fecal microbiota transplantation products. Cell Host Microbe 2020; 27:173–175.
22. Kassam Z, Dubois N, Ramakrishna B, Ling K, Qazi T, Smith M, et al. Donor screening for fecal microbiota transplantation. N Engl J Med 2019; 381:2070–2072.
23. Costello SP, Tucker EC, La Brooy J, Schoeman MN, Andrews JM. Establishing a fecal microbiota transplant service for the treatment of clostridium difficile infection. Clin Infect Dis 2016; 62:908–914.
24. Craven LJ, Nair Parvathy S, Tat-Ko J, Burton JP, Silverman MS. Extended screening costs associated with selecting donors for fecal microbiota transplantation for treatment of metabolic syndrome-associated diseases. Open Forum Infect Dis 2017; 4:ofx243.
25. Kazerouni A, Burgess J, Burns LJ, Wein LM. Optimal screening and donor management in a public stool bank. Microbiome 2015; 3:75.
26. Paramsothy S, Borody TJ, Lin E, Finlayson S, Walsh AJ, Samuel D, et al. Donor recruitment for fecal microbiota transplantation. Inflamm Bowel Dis 2015; 21:1600–1606.
27. Tariq R, Weatherly R, Kammer P, Pardi DS, Khanna S. Donor screening experience for fecal microbiota transplantation in patients with recurrent C. difficile infection. J Clin Gastroenterol 2018; 52:146–150.
28. Robert C, Ribas A, Hamid O, Daud A, Wolchok JD, Joshua AM, et al. Durable complete response after discontinuation of pembrolizumab in patients with metastatic melanoma. J Clin Oncol 2018; 36:1668–1674.
29. Ribas A, Hamid O, Daud A, Hodi FS, Wolchok JD, Kefford R, et al. Association of pembrolizumab with tumor response and survival among patients with advanced melanoma. JAMA 2016; 315:1600–1609.
30. Larkin J, Lao CD, Urba WJ, McDermott DF, Horak C, Jiang J, et al. Efficacy and safety of nivolumab in patients with BRAF V600 mutant and BRAF wild-type advanced melanoma: a pooled analysis of 4 clinical trials. JAMA Oncol 2015; 1:433–440.
31. Curry S, Bogdanovich T, Pakstis D, Schwartz M, Binion D. Fecal microbiota transplantation for treatment of recurrent clostridium difficile infections using recipient-directed donors sero-matched for latent viruses: the University of Pittsburgh Medical Center (UPMC) experience. Open Forum Infect Dis 2016; 3:1200.
32. Babiker A, Ingersoll JM, Adelman MW, Webster AS, Broder KJ, Stittleburg V, et al. Validation of high-sensitivity severe acute respiratory syndrome coronavirus 2 testing for stool-toward the new normal for fecal microbiota transplantation. Clin Transl Gastroenterol 2021; 12:e00363.
33. Ianiro G, Mullish BH, Kelly CR, Kassam Z, Kuijper EJ, Ng SC, et al. Reorganisation of faecal microbiota transplant services during the COVID-19 pandemic. Gut 2020; 69:1555–1563.
34. Ng SC, Chan FKL, Chan PKS. Screening FMT donors during the COVID-19 pandemic: a protocol for stool SARS-CoV-2 viral quantification. Lancet Gastroenterol Hepatol 2020; 5:642–643.
35. Zheng S, Fan J, Yu F, Feng B, Lou B, Zou Q, et al. Viral load dynamics and disease severity in patients infected with SARS-CoV-2 in Zhejiang province, China, January-March 2020: retrospective cohort study. BMJ 2020; 369:m1443.
36. Hyams KC, Krogwold RA, Brock S, Wignall FS, Cross E, Hayes C. Heterosexual transmission of viral hepatitis and cytomegalovirus infection among United States military personnel stationed in the western Pacific. Sex Transm Dis 1993; 20:36–40.
37. Cannon MJ, Schmid DS, Hyde TB. Review of cytomegalovirus seroprevalence and demographic characteristics associated with infection. Rev Med Virol 2010; 20:202–213.
38. Olsson J, Kok E, Adolfsson R, Lovheim H, Elgh F. Herpes virus seroepidemiology in the adult Swedish population. Immun Ageing 2017; 14:10.
39. Xiong G, Zhang B, Huang M-yun, Zhou H, Chen L-Zhen, Feng Q-sheng, et al. Epstein-Barr virus (EBV) infection in Chinese children: a retrospective study of age-specific prevalence. PLoS One 2014; 9:e99857.
40. Wong Y, Meehan MT, Burrows SR, Doolan DL, Miles JJ. Estimating the global burden of Epstein-Barr virus-related cancers. J Cancer Res Clin Oncol 2022; 148:31–46.
41. Nasrallah GK, Dargham SR, Abu-Raddad LJ. Negative epidemiological association between HSV-1 and HSV-2 infections. Heliyon 2019; 5:e02549.
42. Swedan SF, Darabseh A. Seroprevalence of herpes simplex virus types 1 and 2 and correlates of infection in Jordan. Int J STD AIDS 2022; 33:304–310.
43. Malkin JE, Morand P, Malvy D, Ly TD, Chanzy B, de Labareyre C, et al. Seroprevalence of HSV-1 and HSV-2 infection in the general French population. Sex Transm Infect 2002; 78:201–203.
44. Campadelli-Fiume G, Mirandola P, Menotti L. Human herpesvirus 6: an emerging pathogen. Emerg Infect Dis 1999; 5:353–366.
45. Elia F, Villani S, Ambrogi F, Signorini L, Dallari S, Binda S, et al. JC virus infection is acquired very early in life: evidence from a longitudinal serological study. J Neurovirol 2017; 23:99–105.
46. Kolasa M, Hagman S, Verkkoniemi-Ahola A, Airas L, Koivisto K, Elovaara I. Anti-JC virus seroprevalence in a Finnish MS cohort. Acta Neurol Scand 2016; 133:391–397.
47. San Martin H, Balanda M, Vergara N, Valenzuela MA, Cartier L, Ayala S, et al. Human T-lymphotropic virus type 1 and 2 seroprevalence among first-time blood donors in Chile, 2011-2013. J Med Virol 2016; 88:1067–1075.
48. Kaur G, Basu S, Kaur R, Kaur P, Garg S. Patterns of infections among blood donors in a tertiary care centre: a retrospective study. Natl Med J India 2010; 23:147–149.
49. Wanni NHO, Dossary RA, Obeid OE, Qahtani NHA, Siddiqui ZI, El-Badry AA, et al. Seropositivity of syphilis among individuals screened in a tertiary hospital in the Eastern Province of Saudi Arabia. Ann Saudi Med 2021; 41:8–13.
50. Schubert M, Kämpf D, Wahl M, Hofmann S, Girbig M, Jatzwauk L, et al. MRSA point prevalence among health care workers in German rehabilitation centers: a multi-center, cross-sectional study in a non-outbreak setting. Int J Environ Res Public Health 2019; 16:1660.
51. Lin MY, Hayden MK, Lyles RD, Lolans K, Fogg LF, Kallen AJ, et al.; Prevention Epicenters Program, Centers for Disease Control and Prevention. Regional epidemiology of methicillin-resistant staphylococcus aureus among adult intensive care unit patients following state-mandated active surveillance. Clin Infect Dis 2018; 66:1535–1539.
52. Cerrada-Romero C, Berastegui-Cabrera J, Camacho-MartĂnez P, Goikoetxea-Aguirre J, PĂ©rez-Palacios P, SantibĂ¡Ă±ez S, et al. Excretion and viability of SARS-CoV-2 in feces and its association with the clinical outcome of COVID-19. Sci Rep 2022; 12:7397.
53. Heneghan CJ, Spencer EA, Brassey J, PlĂ¼ddemann A, Onakpoya IJ, Evans DH, et al. SARS-CoV-2 and the role of orofecal transmission: a systematic review. F1000Res 2021; 10:231.
54. Smith M, Dai A, Ghilardi G, Amelsberg KV, Devlin SM, Pajarillo R, et al. Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat Med 2022; 28:713–723.
55. Lee K. A, Thomas AM, Bolte LA, Björk JR, de Ruijter LK, Armanini F, et al. Cross-cohort gut microbiome associations with immune checkpoint inhibitor response in advanced melanoma. Nat Med 2022; 28:535–544.
56. Chen J, Zaman A, Ramakrishna B, Olesen SW. Stool banking for fecal microbiota transplantation: methods and operations at a large stool bank. Front Cell Infect Microbiol 2021; 11:622949.
57. DeFilipp Z, Peled JU, Li S, Mahabamunuge J, Dagher Z, Slingerland AE, et al. Third-party fecal microbiota transplantation following allo-HCT reconstitutes microbiome diversity. Blood Adv 2018; 2:745–753.
58. Taur Y, Coyte K, Schluter J, Robilotti E, Figueroa C, Gjonbalaj M, et al. Reconstitution of the gut microbiota of antibiotic-treated patients by autologous fecal microbiota transplant. Sci Transl Med 2018; 10:eaap9489.
59. Kakihana K, Fujioka Y, Suda W, Najima Y, Kuwata G, Sasajima S, et al. Fecal microbiota transplantation for patients with steroid-resistant acute graft-versus-host disease of the gut. Blood 2016; 128:2083–2088.
60. Spindelboeck W, Schulz E, Uhl B, Kashofer K, Aigelsreiter A, Zinke-Cerwenka W, et al. Repeated fecal microbiota transplantations attenuate diarrhea and lead to sustained changes in the fecal microbiota in acute, refractory gastrointestinal graft-versus-host-disease. Haematologica 2017; 102:e210–e213.
61. Xiao F, Tang M, Zheng X, Liu Y, Li X, Shan H. Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterology 2020; 158:1831–1833.e3.
62. Guo M, Tao W, Flavell RA, Zhu S. Potential intestinal infection and faecal-oral transmission of SARS-CoV-2. Nat Rev Gastroenterol Hepatol 2021; 18:269–283.
63. Peiro-Mestres A, Fuertes I, CamprubĂ-Ferrer D, Marcos MA, Vilella A, Navarro M, et al. Frequent detection of monkeypox virus DNA in saliva, semen, and other clinical samples from 12 patients, Barcelona, Spain, May to June 2022. Euro Surveill 2022; 27:2200503.
64. Ashraf MF, Tageldin O, Nassar Y, Batool A. Fecal microbiota transplantation in patients with recurrent clostridium difficile infection: a four-year single-center retrospective review. Gastroenterology Res 2021; 14:237–243.