Hereditary gastrointestinal cancer is associated with molecular and neoplastic precursors which have revealed much about sporadic carcinogenesis. Therefore, an appreciation of constitutional and somatic events linked to these syndromes have provided a useful model for the development of risk models and preventative strategies. In this review, we focus of two of the best characterized syndromes, Lynch syndrome (LS) and familial adenomatous polyposis (FAP). Our understanding of the neoplasia-immune interaction in LS has contributed to the development of immune mediated therapies including cancer preventing vaccines and immunotherapy for cancer precursors. Chemoprevention in LS with aspirin and nonsteroidal anti-inflammatory drugs has also translated into clinical cancer, however the efficacy of such agents in FAP remains elusive when cancer is applied as an endpoint in trials rather than the use of ‘indirect’ endpoints such as polyp burden, and requires further elucidation of biological mechanisms in FAP. Finally, we review controversies in gastrointestinal surveillance for LS and FAP, including limitations and opportunities of upper and lower gastrointestinal endoscopy in the prevention and early detection of cancer.

Cancer is thought to be a multistep evolutionary process arising from a single cell, acquiring genomic alterations, which provide a fitness advantage (1, 2). Syndromic constitutional pathogenic variants (PV) predispose individuals to a high risk of developing cancer, providing an opportunity to identify and explore precancerous lesions in these patients. Of the cancer susceptibility syndromes affecting the gastrointestinal tract, the two best defined syndromes are Lynch syndrome (LS) and familial adenomatous polyposis (FAP). Studying these groups on regular cancer surveillance provides insight into the tumor biology, potential avenues for prevention, and their effectiveness. In this article, we will review some of the lessons that can be learned from these conditions which may afford opportunities for cancer prevention.

FAP is a rare dominantly inherited syndrome, the hallmark of which is the development of up to hundreds or thousands of colorectal adenomas and almost inevitable development of colorectal cancer without intervention. Constitutional PV's in the tumor suppressor gene adenomatous polyposis coli (APC) result in constitutive activation of the Wnt signaling pathway through deregulation of β-catenin, causing downstream effects on proliferation and differentiation within colonic crypts. Somatic APC variants are also found in more than 80% of sporadic colorectal tumors. Given its well defined phenotype FAP has been considered a good model for the study of colorectal cancer and chemoprevention. Due to the ubiquitous expression of APC, it is not surprising that FAP is a multisystem disorder and there are other precancerous lesions to consider, for which intervention may be required.

LS is an autosomal dominant inherited condition which increases the lifetime risk of colorectal cancer, as well as other predominantly epithelial cancers, and affects 1 in 400 adults (3, 4). It is defined by the presence of a constitutional PV in one of the DNA replication mismatch repair (MMR) genes. LS-related cancer diagnoses are common in younger people below national screening ages, with a significant impact on quality-adjusted life years gained through early diagnosis and prevention. Therefore, interventions are applied to try and mitigate these risks, which include regular colonoscopy, chemoprophylaxis and preventative gynecologic surgery (5). Thorough screening programs throughout patients’ lifetimes allow for multiple time-point studies of the evolution of LS-associated cancers from pre-dysplasia (normal biopsies) through to early and late dysplasia (adenocarcinoma), illuminating the otherwise unseen precancerous stage of lesion development in sporadic cancers. Recent cancer prevention and early detection innovations have moved towards the integration of molecular knowledge and risk stratification profiles, to allow for a more accurate characterization of at-risk individuals (6).

Mismatch repair deficiency (dMMR) drives mutagenesis and genomic instability in LS-associated cancer, along with approximately 15% of sporadic colorectal cancer. dMMR is not itself an initiator of oncogenesis alone, but instead accelerates mutation accumulation and therefore the chances of acquiring a driver mutation, a phenomenon termed hypermutator phenotype.

Compelling data have been published relating to premalignant molecular events in the context of LS colorectal cancer, including the identification of dMMR but morphologically nonneoplastic colonic crypts (7, 8). These crypts exhibit MMR gene variants and are likely precursors to malignancy, but also undergo a high rate of regression under immune surveillance. Other evidence that dMMR is an early event in LS carcinogenesis includes data which indicate that 77% of adenomas are dMMR, and the heterogeneity of somatic driver mutations in LS cancers other than dMMR (9). Indeed, Ahadova and colleagues described how known colorectal cancer mutations such as KRAS and APC mutations commonly occur after the onset of dMMR in LS (Fig. 1). Together this suggests dMMR clones evoke high negative selective pressures on them by the immune system; a mechanism that could potentially be harnessed and heightened by preventative medicine.

Figure 1.

Model of the potential pathway towards cancer in LS. Somatic loss of the functional wild-type MMR allele is thought to be the initiating event in LS dysplasia, causing a hypermutator phenotype which subsequently acquires mutations providing further fitness advantage, such as immune escape and oncogenic driver mutations.

Figure 1.

Model of the potential pathway towards cancer in LS. Somatic loss of the functional wild-type MMR allele is thought to be the initiating event in LS dysplasia, causing a hypermutator phenotype which subsequently acquires mutations providing further fitness advantage, such as immune escape and oncogenic driver mutations.

Close modal

The time required for cancer development in LS has been estimated to be much shorter than in sporadic colorectal cancer: the time interval between a clear colonoscopy and later carcinoma formation in LS is around 2 to 3 years (10) whereas estimations of sporadic cancer evolution suggest it could take decades to evolve (11), with incidences of sporadic cancer within 3 years of a clear colonoscopy being quite rare (12). This acceleration of cancer onset is attributed to bypassing the need for two loss of function events in each of the MMR alleles, leading to an increase in mutational rate and therefore likelihood of oncogenic mutations (13). The shortened timeframe of cancer evolution in LS allows for a timelier investigation of the processes needed for cells to overcome for malignancy. The information from LS studies may also be translated to understanding the longer sporadic colorectal cancer development.

The identification of ‘submucosal’ colorectal cancer is a phenomenon rarely observed outside the context of LS. It may account for only a small number of colorectal cancer in the context of LS, but nevertheless suggests that some cancers could arise via a non-adenoma precursor route (Fig. 2; ref. 14). Colonoscopy may have limited efficacy in the prevention of such colorectal cancers, and require augmentation in its approach, or goals more focused on early diagnosis than prevention. This pathway of progression seems to be more common in MLH1 and MSH2 variant carriers (15) and less common in PMS2-associated colorectal cancer, an observation that may reflect the minimal risk of cancers in PMS2 carriers under surveillance. It has been shown that CTNNB1 and TP53 mutations occur more commonly in tumors lacking evidence of non-flat morphology polypoid growth (9).

Figure 2.

Submucosal colorectal lesions in an MLH1 germline LS patient. Single adenoma with (A) low grade dysplasia and (B) high-grade dysplasia, with crypts in the submucosa on the right. C, Ileocolic anastomosis, (D) splenic flexure, and (E) polypectomy.

Figure 2.

Submucosal colorectal lesions in an MLH1 germline LS patient. Single adenoma with (A) low grade dysplasia and (B) high-grade dysplasia, with crypts in the submucosa on the right. C, Ileocolic anastomosis, (D) splenic flexure, and (E) polypectomy.

Close modal

Unexpectedly high rates of post-colonoscopy colorectal cancer have been reported, with cumulative incidences up to 45% in MLH1 carriers, for example, in comparison of the 9.3% rates reported in population-based screening programs. It is important to note that none of these studies considered the potential impact of quality of colonoscopy on this potential failure of colonoscopy surveillance. These data have led to the generation of the hypothesis that post-colonoscopy colorectal cancer in LS arises from an “invisible” subepithelial lesion that progresses directly to colorectal cancer. However, the true proportion of submucosal versus mucosal tumors is unknown (16).

Tumor mutational signatures may indicate a genetic predisposition that underlies the cancer, and facilitate personalized cancer therapy and prevention, by integrating the somatic mutation landscape within a single tumor to identify somatic molecular patterns associated with distinct oncogenic pathways (17). In addition, appreciation of early somatic events in hereditary colorectal cancer precursors including adenomas may contribute towards prediction models for progression of patients undergoing longitudinal endoscopic surveillance (18).

Mismatch variants accumulate in dMMR cancers resulting in hypermutated tumors with somatic mutations, including insertion/deletion mutations within repetitive sequences which are susceptible to replication errors, including in exons (coding microsatellites, cMS). These indels promote translational frameshifts, which generate truncated frameshift peptide (FSP)-encoding neoproteins (19). Several studies have identified a large spectrum of genes affected by such frameshift mutations, demonstrating that indel mutations affecting key tumor suppressors, such as the TGFBR2, are enriched in dMMR cancers. Specific dMMR-related FSP neoantigens can encompass neo-epitopes completely unknown to the host's immune system. This mechanism is commonly considered to be responsible for the high immunogenicity of dMMR cancers, demonstrated by several studies showing dense local immune cell infiltration and reactivity to FSPs. Nonneoplastic dMMR colonic crypts are seen in healthy LS carriers, suggesting that the healthy colon of patients with LS is itself a key source of immunogenic FSPs that serve to auto-vaccinate such patients and suppress dMMR-induced carcinogenesis (7).

Vaccination with recurrent FSPs that are shared by multiple dMMR tumors of different patients is a promising approach to boost immune surveillance of dMMR precancerous cell clones, and potentially immune-mediated regression of subclinical dMMR tumors for effective immunoprevention. A phase IIa clinical trial in patients with a history of dMMR colorectal cancer has demonstrated the safety and immunologic efficacy of a trivalent recurrent FSP-based vaccine (19). Whether recurrent FSP vaccination can reduce LS or sporadic dMMR tumor burden or prolong patient survival is unknown however.

A more precise understanding of the mechanisms by which LS-associated carcinogenesis escapes immune surveillance may facilitate the translation of such discoveries into immune-based cancer prevention, for example by elucidating the role of HLA genotypes. Early data from mouse models suggest this is a potentially fruitful mechanism in combination with NSAIDs, with early studies in human subjects indicating the safety of LS vaccines (19, 20).

In sporadic colorectal cancer, correlations have been made between gut microorganisms, intestinal barrier function and inflammation. There are few data to indicate a ‘microbiome signature’ in hereditary colorectal cancer syndromes (21). There appears to be an interaction between MMR and TGFBR2 inactivation in inflammation-associated colon tumorigenesis (22). However, the dominant characteristic thus far of the microbiome in LS relates to previous colorectal resection (21).

In FAP, biofilms including Escherichia coli and Bacteroides fragilis have been observed penetrating the mucous barrier, indicating that the intestinal barrier function is compromised (23). There are data suggesting impaired cellular immunity and tumor surveillance in FAP. This implies that mucosal immune dysfunction may contribute to carcinogenesis in this predominantly genetically driven condition (24).

Microbiome signatures from average risk screening populations may augment existing population screening tools, including the fecal immunochemical test. This is being explored as part of an existing multicenter trial of people with LS in the UK (25). Other potential biomarkers are being evaluated in patients with LS, with the advantage that the benefits in such populations may be more tangible in a shorter timeframe, before translation of other predominantly high-risk populations. These include organ-specific biomarkers (e.g., urinary biomarkers for endometrial or urothelial cancer and fecal or plasma genomic markers for colorectal cancer), as well as non–organ-specific biomarkers including cell free DNA from plasma (26–29).

Although much focus is on the cancer risk in the large bowel in both FAP and LS, both carry an increased risk of cancer relating to the upper gastrointestinal (UGI) tract.

In FAP, there are emerging data regarding gastric adenomas and indeed an increased risk of gastric cancer in FAP (30–33), but these entities in FAP are yet to be well defined in terms of life-time risk, pathways to cancer and speed of cancer development; furthermore the differences in gastric cancer risk and pathways to cancer between the Western World and Asia make interpreting data in FAP even more challenging. It is also clear that there is an increased risk for gastric and duodenal cancer in patients with LS. However, the current data are difficult to interpret due to ascertainment bias and the true lifetime risks are not well established. Upper gastrointestinal tract surveillance in LS is variably recommended (5, 34) and this variability may relate to few data regarding the incidence, natural history or intervention outcomes for premalignant lesions. Therefore, it is difficult to extrapolate any of these data to sporadic disease.

Sporadic adenomas in the duodenum are rare, with a prevalence as low as 0.03% in those referred for diagnostic upper gastrointestinal endoscopy (35). They are usually sited at the level of, or just distal to, the ampulla of Vater. In FAP, duodenal adenomas are common; indeed, an inherited polyposis syndrome may account for 60% or more of those patients in whom a duodenal adenoma is found (36). The distribution of adenomas in FAP mirrors that of sporadic duodenal adenomas, with a propensity to develop in the peri-ampullary area (37).

In FAP, the lifetime risks of developing one or more adenomas approaches 100% (38) and of developing duodenal cancer is 5% to 10%therefore it is an area which seems ripe for study to better understand the sporadic counterpart. A staging system has been adopted (Table 1). Prospective data from a number of studies have shown that there is slow progression of all stages of duodenal disease which infrequently develop cancer (38–40). Understanding this natural history is useful in terms of understanding if and when to intervene in those with sporadic duodenal adenomas, or for studying the effect of any intervention.

Table 1.

Spigelman classification of duodenal disease in FAP (66).

Findings at duodenoscopy1 point2 points3 points
Number of adenomas 1–4 5–20 >20 
Size (mm) 1–4 5–10 >10 
Histology* Tubular Tubulovillous Villous 
Dysplasia* Low grade NA High grade 
Findings at duodenoscopy1 point2 points3 points
Number of adenomas 1–4 5–20 >20 
Size (mm) 1–4 5–10 >10 
Histology* Tubular Tubulovillous Villous 
Dysplasia* Low grade NA High grade 

Stage 0 = score 0.

Stage I = score 1–4.

Stage II – score 5–7.

Stage III = score 7–8.

Stage IV = score 9–12.

Bile is a likely aetiological factor in duodenal adenoma development. The distribution of adenomas within the duodenum mimics exposure to bile in both sporadic and FAP duodenal adenomas. Bile from FAP and non-FAP patients has similar mutagenicity, however, the composition of bile relating to bile acids does appear to differ (41, 42). DNA adduct formation, as a measure DNA carcinogen exposure, is increased in FAP versus controls (43–45). Furthermore, the dietary administration of unconjugated bile, in a mouse model of FAP, increases duodenal tumor burden (46). Further studies to better define the etiology of duodenal adenoma in FAP, may well have direct benefits in terms of understanding and managing sporadic disease and directing new therapies.

Exploring chemoprevention in inherited conditions with a high cancer risk may be helpful as a model for sporadic cancer prevention. One of the difficulties in such studies is establishing what is an appropriate primary endpoint to measure. A reduction in cancer risk is the most important endpoint. In LS, with its lack of polyposis phenotype, then cancer has been used appropriately as the endpoint to be measured. However, in FAP, where there are evidence-based interventions to reduce cancer risk in the large bowel (namely prophylactic surgery) and where cancer risk in the duodenum remains uncommon, a number of surrogate endpoints have been used, however there remains much debate as to whether these endpoints in FAP are clinically meaningful and appropriate.

Aspirin and nonsteroidal anti-inflammatory drugs have been the most widely studied chemoprevention drugs in LS, FAP, and indeed, sporadic colorectal cancer. In CAPP2, patients with LS were assigned to receive 600-mg aspirin daily or placebo. The primary endpoint was development of colorectal cancer. The initial report was of significant benefit (47), however longer-term follow up outcomes demonstrate a reduction in colorectal cancer risk in those receiving aspirin [HR, 0.65; 95% confidence interval (CI), 0.43–0.97; P = 0.035] for aspirin versus placebo. These results reflects the growing data on the benefit of aspirin in prevention of sporadic colorectal cancer (48). It is interesting however to note that the timeframe before the benefit is shorter in LS than sporadic colorectal cancer, where benefit may be seen after 10 years (49). This difference likely relates to the rates of carcinogenesis between LS and sporadic colorectal cancer (49). Studies of aspirin in FAP have yielded conflicting results, likely due to the methodologies used (Table 2)

Table 2.

Chemoprophyllaxis trials in hereditary colorectal cancer (67–71).

AuthorDrugStudy designCohort sizeEndpointBenefit
Steinbach Celecoxib Placebo RCT 77 Polyp burden Yes 
West Eicosapaentanoic acid Placebo RCT 55 Polyp burden Yes 
Lynch CXB/DFMO vs. CXB RCT 112 Polyp burden No 
Burn Aspirin ± starch RCT 206 Polyp burden No 
Ishikawa Aspirin ± mesalazine RCT 104 Polyp recurrence Yes 
AuthorDrugStudy designCohort sizeEndpointBenefit
Steinbach Celecoxib Placebo RCT 77 Polyp burden Yes 
West Eicosapaentanoic acid Placebo RCT 55 Polyp burden Yes 
Lynch CXB/DFMO vs. CXB RCT 112 Polyp burden No 
Burn Aspirin ± starch RCT 206 Polyp burden No 
Ishikawa Aspirin ± mesalazine RCT 104 Polyp recurrence Yes 

Sulindac and eflornithine was studied against either single agent alone, initially no difference in outcomes for the lower gastrointestinal tract was reported (50), but a post hoc analysis of participants with at least a partial intact lower gastrointestinal tract showed a statistically significant reduction in disease progression and need for surgery (once censored for polypectomy >10 mm; ref. 51). The combination of sulindac with erlotinib (52) and single agent weekly erlotinib (53) lead to a significant reduction in polyp burden after 6 months of treatment compared with placebo adverse events were common, limiting their potential as treatment in an otherwise well population, in these combination therapy studies.

Other agents have been trialled in FAP (Table 2), all of which have similar methodologic flaws; being short term and lacking a hard endpoint of cancer and using surrogate endpoints, the validity of which have to be questioned. This is especially important given there are a number of reports of cancer arising whilst on chemoprevention (54–57). Currently there are no data to support any agent preventing colorectal cancer in FAP, along with prevention of surgery data remaining weak with inadequate follow up.

Studies have addressed chemoprevention in the setting of duodenal polyposis in FAP. Sulindac has been studied in several randomized and non-randomized settings (58–61) and in combination with eflornithine (50). Given the possible role of bile and that bile-induced DNA adduct formation is pH-sensitive, ranitidine and the bile salt sequestrant ursodeoxycholic acid have also been investigated in FAP (62, 63). All failed to demonstrate regression of duodenal disease. Furthermore, all studies except the combination study were small and short-term.

Combined treatment with sulindac and erlotinib (52) and a phase II study with weekly erlotinib (53) have shown statistical benefit in terms of polyp burden but adverse events were unacceptably high. Celecoxib treatment (800 mg/day) leads to a nonsignificant decrease in polyp burden, although subgroup analysis showed a significant decrease in polyp burden in those with Spigelman stage III or IV disease (64).

In summary, no agent has been shown to reduce long-term duodenal cancer risk and none have been shown to reduce the need for major resectional surgery, which are the most clinically meaningful endpoints in FAP. With a better understanding of the etiology of duodenal adenomas, more targeted agents could be considered as a chemoprevention strategy to study.

In LS there is less data regarding chemoprevention in the upper gastrointestinal tract. In the CAPP2 study, a reduction in risk for extra-colonic LS-related cancers, including gastric and duodenal cancers, was not observed. However, a protective effect of resistant starch against non–colorectal cancer LS cancers [incidence rate ratios (IRR), 0.52; 95% CI, 0.32–0.84; P = 0.0075] was noted (65).

It is clear that for chemoprevention studies to be appropriately designed, there needs to be an understanding of tumor biology to dictate timeframes required for follow up. Robust clinically meaningful endpoints are required; cancer as an endpoint however may not be possible, as the size of study to show benefit and the duration may not be feasible for new agents with a role in cancer prevention in the setting a non-accelerated cancer pathway. These problems may limit the utility of FAP as a model to study sporadic colorectal cancer chemoprevention.

Hereditary syndromes provide an opportunity to study early precancer evolution surveillance and interventions to prevent cancer. Understanding genotype and phenotype in colorectal cancer susceptibility syndromes allows for customized clinical trials in preventive medicine to be highly effective in their outcomes. The increasing knowledge of precancer lesions and their biology may aid novel strategies for individualized prevention. It is essential to consider the adverse effects of such interventions when weighed against quality of life, particularly considering the age at which medication may begin. Exciting advancements are being made in the field of cancer vaccination against LS colorectal cancer, however until trials show their validity the recommended surveillance protocols must be upheld. Extrapolating information from these hereditary colorectal cancer studies can also help pinpoint how certain sporadic cancers evolve and offer opportunities for cancer prevention.

No disclosures were reported.

1.
Nowell
PC
.
The clonal evolution of tumor cell populations
.
Science
1976
;
194
:
23
8
.
2.
Greaves
M
,
Maley
CC
.
Clonal evolution in cancer
.
Nature
2012
;
481
:
306
13
.
3.
Patel
AP
,
Wang
M
,
Fahed
AC
,
Mason-Suares
H
,
Brockman
D
,
Pelletier
R
, et al
.
Association of rare pathogenic DNA variants for familial hypercholesterolemia, hereditary breast and ovarian cancer syndrome, and Lynch syndrome with disease risk in adults according to family history
.
JAMA Netw Open
2020
;
3
:
e203959
.
4.
Grzymski
JJ
,
Elhanan
G
,
Morales Rosado
JA
,
Smith
E
,
Schlauch
KA
,
Read
R
, et al
.
Population genetic screening efficiently identifies carriers of autosomal dominant diseases
.
Nat Med
2020
;
26
:
1235
9
.
5.
Monahan
KJ
,
Bradshaw
N
,
Dolwani
S
,
Desouza
B
,
Dunlop
MG
,
East
JE
, et al
.
Guidelines for the management of hereditary colorectal cancer from the British Society of Gastroenterology (BSG)/Association of Coloproctology of Great Britain and Ireland (ACPGBI)/United Kingdom Cancer Genetics Group (UKCGG)
.
Gut
2020
;
69
:
411
44
.
6.
Llach
J
,
Pellisé
M
,
Monahan
K
.
Lynch syndrome; towards more personalized management?
Best Pract Res Clin Gastroenterol
2022
;
58–59
:
101790
.
7.
Kloor
M
,
Huth
C
,
Voigt
AY
,
Benner
A
,
Schirmacher
P
,
von Knebel Doeberitz
M
, et al
.
Prevalence of mismatch repair–deficient crypt foci in Lynch syndrome: a pathological study
.
Lancet Oncol
2012
;
13
:
598
606
.
8.
Brand
RE
,
Dudley
B
,
Karloski
E
,
Das
R
,
Fuhrer
K
,
Pai
RK
, et al
.
Detection of DNA mismatch repair–deficient crypts in random colonoscopic biopsies identifies Lynch syndrome patients
.
Fam Cancer
2020
;
19
:
169
75
.
9.
Ahadova
A
,
Gallon
R
,
Gebert
J
,
Ballhausen
A
,
Endris
V
,
Kirchner
M
, et al
.
Three molecular pathways model colorectal carcinogenesis in Lynch syndrome
.
Int J Cancer
2018
;
143
:
139
50
.
10.
Edelstein
DL
,
Axilbund
J
,
Baxter
M
,
Hylind
LM
,
Romans
K
,
Griffin
CA
, et al
.
Rapid development of colorectal neoplasia in patients with Lynch syndrome
.
Clin Gastroenterol Hepatol
2011
;
9
:
340
3
.
11.
Jones
S
,
Chen
W-D
,
Parmigiani
G
,
Diehl
F
,
Beerenwinkel
N
,
Antal
T
, et al
.
Comparative lesion sequencing provides insights into tumor evolution
.
Proc Natl Acad Sci USA
2008
;
105
:
4283
8
.
12.
Schoen
RE
,
Pinsky
PF
,
Weissfeld
JL
,
Yokochi
LA
,
Church
T
,
Laiyemo
AO
, et al
.
Colorectal-cancer incidence and mortality with screening flexible sigmoidoscopy
.
N Engl J Med
2012
;
366
:
2345
57
.
13.
von Loga
K
,
Woolston
A
,
Punta
M
,
Barber
LJ
,
Griffiths
B
,
Semiannikova
M
, et al
.
Extreme intratumor heterogeneity and driver evolution in mismatch repair–deficient gastroesophageal cancer
.
Nat Commun
2020
;
11
:
139
.
14.
Ahadova
A
,
Seppälä
TT
,
Engel
C
,
Gallon
R
,
Burn
J
,
Holinski-Feder
E
, et al
.
The “unnatural” history of colorectal cancer in Lynch syndrome: lessons from colonoscopy surveillance
.
Int J cancer
2021
;
148
:
800
11
.
15.
Engel
C
,
Ahadova
A
,
Seppälä
TT
,
Aretz
S
,
Bigirwamungu-Bargeman
M
,
Bläker
H
, et al
.
Associations of pathogenic variants in MLH1, MSH2, and MSH6 with risk of colorectal adenomas and tumors and with somatic mutations in patients with Lynch syndrome
.
Gastroenterology
2020
;
158
:
1326
33
.
16.
Bajwa-Ten Broeke
SW
,
Ballhausen
A
,
Ahadova
A
,
Suerink
M
,
Bohaumilitzky
L
,
Seidler
F
, et al
.
The coding microsatellite mutation profile of PMS2-deficient colorectal cancer
.
Exp Mol Pathol
2021
;
122
:
104668
.
17.
Tate
JG
,
Bamford
S
,
Jubb
HC
,
Sondka
Z
,
Beare
DM
,
Bindal
N
, et al
.
COSMIC: the catalogue of somatic mutations in cancer
.
Nucleic Acids Res
2018
;
47
:
D941
7
.
18.
Swinyard
O
,
Baker
A-M
,
Kimberly
C
,
Jansen
M
,
Monahan
KJ
,
Graham
TA
.
Coevolution of mismatch repair loss and the immune response in Lynch syndrome
.
Insight
2022
;
21
:
557
636
.
19.
Gebert
J
,
Gelincik
O
,
Oezcan-Wahlbrink
M
,
Marshall
JD
,
Hernandez-Sanchez
A
,
Urban
K
, et al
.
Recurrent frameshift neoantigen vaccine elicits protective immunity with reduced tumor burden and improved overall survival in a Lynch syndrome mouse model
.
Gastroenterology
2021
;
161
:
1288
302
.
20.
Kloor
M
,
Reuschenbach
M
,
Pauligk
C
,
Karbach
J
,
Rafiyan
M-R
,
Al-Batran
S-E
, et al
.
A frameshift peptide neoantigen-based vaccine for mismatch repair–deficient cancers: a phase I/IIa clinical trial
.
Clin Cancer Res
2020
;
26
:
4503
10
.
21.
Yan
Y
,
Drew
DA
,
Markowitz
A
,
Lloyd-Price
J
,
Abu-Ali
G
,
Nguyen
LH
, et al
.
Structure of the mucosal and stool microbiome in Lynch syndrome
.
Cell Host Microbe
2020
;
27
:
585
600
.
22.
Tosti
E
,
Almeida
AS
,
Tran
TTT
,
Barbachan
E
Silva
M
,
Broin
, et al
.
Loss of MMR and TGFBR2 increases the susceptibility to microbiota-dependent inflammation-associated colon cancer
.
Cell Mol Gastroenterol Hepatol
2022
;
14
:
693
717
.
23.
Dejea
CM
,
Fathi
P
,
Craig
JM
,
Boleij
A
,
Taddese
R
,
Geis
AL
, et al
.
Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria
.
Science
2018
;
359
:
592
7
.
24.
Noble
A
,
Durant
L
,
Dilke
SM
,
Man
R
,
Martin
I
,
Patel
R
, et al
.
Altered mucosal immune-microbiota interactions in familial adenomatous polyposis
.
Clin Transl Gastroenterol
2022
;
13
:
e00428
.
25.
Lincoln
A
,
Benton
S
,
Piggott
C
,
North
BV
,
Rigney
J
,
Young
C
, et al
.
Exploring the utility and acceptability of fecal immunochemical testing (FIT) as a novel intervention for the improvement of colorectal cancer (CRC) surveillance in individuals with lynch syndrome (FIT for Lynch study): a single-arm, prospective, multicenter, non-randomized study
.
BMC Cancer
2022
;
22
:
1144
.
26.
Njoku
K
,
Chiasserini
D
,
Jones
ER
,
Barr
CE
,
O'Flynn
H
,
Whetton
AD
, et al
.
Urinary biomarkers and their potential for the noninvasive detection of endometrial cancer
.
Front Oncol
2020
;
10
:
559016
.
27.
van Liere
ELSA
,
de Boer
NKH
,
Dekker
E
,
van Leerdam
ME
,
de Meij
TGJ
,
Ramsoekh
D
.
Systematic review: non-endoscopic surveillance for colorectal neoplasia in individuals with Lynch syndrome
.
Aliment Pharmacol Ther
2022
;
55
:
778
88
.
28.
Hitchins
MP
,
Vogelaar
IP
,
Brennan
K
,
Haraldsdottir
S
,
Zhou
N
,
Martin
B
, et al
.
Methylated SEPTIN9 plasma test for colorectal cancer detection may be applicable to Lynch syndrome
.
BMJ Open Gastroenterol
2019
;
6
:
e000299
.
29.
Ballester
V
,
Taylor
WR
,
Slettedahl
SW
,
Mahoney
DW
,
Yab
TC
,
Sinicrope
FA
, et al
.
Novel methylated DNA markers accurately discriminate Lynch syndrome associated colorectal neoplasia
.
Epigenomics
2020
;
12
:
2173
87
.
30.
Mankaney
G
,
Leone
P
,
Cruise
M
,
LaGuardia
L
,
O'Malley
M
,
Bhatt
A
, et al
.
Gastric cancer in FAP: a concerning rise in incidence
.
Fam Cancer
2017
;
16
:
371
6
.
31.
Walton
S-J
,
Frayling
IM
,
Clark
SK
,
Latchford
A
.
Gastric tumors in FAP
.
Fam Cancer
2017
;
16
:
363
9
.
32.
Martin
I
,
Roos
VH
,
Anele
C
,
Walton
S-J
,
Cuthill
V
,
Suzuki
N
, et al
.
Gastric adenomas and their management in familial adenomatous polyposis
.
Endoscopy
2021
;
53
:
795
801
.
33.
Leone
PJ
,
Mankaney
G
,
Sarvapelli
S
,
Abushamma
S
,
Lopez
R
,
Cruise
M
, et al
.
Endoscopic and histologic features associated with gastric cancer in familial adenomatous polyposis
.
Gastrointest Endosc
2019
;
89
:
961
8
.
34.
Seppälä
TT
,
Latchford
A
,
Negoi
I
,
Sampaio Soares
A
,
Jimenez-Rodriguez
R
,
Sánchez-Guillén
L
, et al
.
European guidelines from the EHTG and ESCP for Lynch syndrome: an updated third edition of the Mallorca guidelines based on gene and gender
.
Br J Surg
2021
;
108
:
484
98
.
35.
Jung
SH
,
Chung
WC
,
Kim
EJ
,
Kim
SH
,
Paik
CN
,
Lee
BI
, et al
.
Evaluation of non-ampullary duodenal polyps: comparison of nonneoplastic and neoplastic lesions
.
World J Gastroenterol
2010
;
16
:
5474
80
.
36.
Johnson
MD
,
Mackey
R
,
Brown
N
,
Church
J
,
Burke
C
,
Walsh
RM
.
Outcome based on management for duodenal adenomas: sporadic versus familial disease
.
J Gastrointest Surg
2010
;
14
:
229
35
.
37.
Domizio
P
,
Talbot
IC
,
Spigelman
AD
,
Williams
CB
,
Phillips
RK
.
Upper gastrointestinal pathology in familial adenomatous polyposis: results from a prospective study of 102 patients
.
J Clin Pathol
1990
;
43
:
738
43
.
38.
Bülow
S
,
Björk
J
,
Christensen
IJ
,
Fausa
O
,
Järvinen
H
,
Moesgaard
F
, et al
.
Duodenal adenomatosis in familial adenomatous polyposis
.
Gut
2004
;
53
:
381
6
.
39.
Burke
CA
,
Beck
GJ
,
Church
JM
,
van Stolk
RU
.
The natural history of untreated duodenal and ampullary adenomas in patients with familial adenomatous polyposis followed in an endoscopic surveillance program
.
Gastrointest Endosc
1999
;
49
:
358
64
.
40.
Groves
CJ
,
Saunders
BP
,
Spigelman
AD
,
Phillips
RKS
.
Duodenal cancer in patients with familial adenomatous polyposis (FAP): results of a 10-year prospective study
.
Gut
2002
;
50
:
636
41
.
41.
Spigelman
AD
,
Crofton-Sleigh
C
,
Venitt
S
,
Phillips
RK
.
Mutagenicity of bile and duodenal adenomas in familial adenomatous polyposis
.
Br J Surg
1990
;
77
:
878
81
.
42.
Spigelman
AD
,
Owen
RW
,
Hill
MJ
,
Phillips
RK
.
Biliary bile acid profiles in familial adenomatous polyposis
.
Br J Surg
1991
;
78
:
321
5
.
43.
Spigelman
AD
,
Scates
DK
,
Venitt
S
,
Phillips
RK
.
DNA adducts, detected by 32P-postlabelling, in the foregut of patients with familial adenomatous polyposis and in unaffected controls
.
Carcinogenesis
1991
;
12
:
1727
32
.
44.
Scates
DK
,
Spigelman
AD
,
Phillips
RK
,
Venitt
S
.
DNA adducts detected by 32P-postlabelling, in the intestine of rats given bile from patients with familial adenomatous polyposis and from unaffected controls
.
Carcinogenesis
1992
;
13
:
731
5
.
45.
Scates
DK
,
Spigelman
AD
,
Nugent
KP
,
Phillips
RK
,
Venitt
S
.
DNA adducts, detected by 32P-postlabelling, in DNA treated in vitro with bile from patients with familial adenomatous polyposis and from unaffected controls
.
Carcinogenesis
1993
;
14
:
1107
10
.
46.
Mahmoud
NN
,
Dannenberg
AJ
,
Bilinski
RT
,
Mestre
JR
,
Chadburn
A
,
Churchill
M
, et al
.
Administration of an unconjugated bile acid increases duodenal tumors in a murine model of familial adenomatous polyposis
.
Carcinogenesis
1999
;
20
:
299
303
.
47.
Burn
J
,
Gerdes
A-M
,
Macrae
F
,
Mecklin
J-P
,
Moeslein
G
,
Olschwang
S
, et al
.
Long-term effect of aspirin on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomized controlled trial
.
Lancet
2011
;
378
:
2081
7
.
48.
Qiao
Y
,
Yang
T
,
Gan
Y
,
Li
W
,
Wang
C
,
Gong
Y
, et al
.
Associations between aspirin use and the risk of cancers: a meta-analysis of observational studies
.
BMC Cancer
2018
;
18
:
288
.
49.
Cook
NR
,
Lee
I-M
,
Zhang
SM
,
Moorthy
MV
,
Buring
JE
.
Alternate-day, low-dose aspirin and cancer risk: long-term observational follow-up of a randomized trial
.
Ann Intern Med
2013
;
159
:
77
85
.
50.
Burke
CA
,
Dekker
E
,
Lynch
P
,
Samadder
NJ
,
Balaguer
F
,
Hüneburg
R
, et al
.
Eflornithine plus sulindac for prevention of progression in familial adenomatous polyposis
.
N Engl J Med
2020
;
383
:
1028
39
.
51.
Balaguer
F
,
Stoffel
EM
,
Burke
CA
,
Dekker
E
,
Samadder
NJ
,
Van Cutsem
E
, et al
.
Combination of sulindac and eflornithine delays the need for lower gastrointestinal surgery in patients with familial adenomatous polyposis: post hoc analysis of a randomized clinical trial
.
Dis Colon Rectum
2022
;
65
:
536
45
.
52.
Samadder
NJ
,
Neklason
DW
,
Boucher
KM
,
Byrne
KR
,
Kanth
P
,
Samowitz
W
, et al
.
Effect of sulindac and erlotinib vs placebo on duodenal neoplasia in familial adenomatous polyposis: a randomized clinical trial
.
JAMA
2016
;
315
:
1266
75
.
53.
Samadder
NJ
,
Foster
N
,
McMurray
RP
,
Burke
CA
,
Stoffel
E
,
Kanth
P
, et al
.
Phase II trial of weekly erlotinib dosing to reduce duodenal polyp burden associated with familial adenomatous polyposis
.
Gut
2023
;
72
:
256
63
.
54.
Tonelli
F
,
Valanzano
R
,
Messerini
L
,
Ficari
F
.
Long-term treatment with sulindac in familial adenomatous polyposis: is there an actual efficacy in prevention of rectal cancer?
J Surg Oncol
2000
;
74
:
15
20
.
55.
Thorson
AG
,
Lynch
HT
,
Smyrk
TC
.
Rectal cancer in FAP patient after sulindac
.
Lancet
1994
;
343
:
180
.
56.
Lynch
HT
,
Thorson
AG
,
Smyrk
T
.
Rectal cancer after prolonged sulindac chemoprevention: a case report
.
Cancer
1995
;
75
:
936
8
.
57.
Utech
M
,
Brüwer
M
,
Buerger
H
,
Tübergen
D
,
Senninger
N
.
Rectal carcinoma in a patient with familial adenomatous polyposis coli after colectomy with ileorectal anastomosis and consecutive chemoprevention with sulindac suppositories
.
Chirurg
2002
;
73
:
855
8
.
58.
Winde
G
,
Schmid
KW
,
Brandt
B
,
Müller
O
,
Osswald
H
.
Clinical and genomic influence of sulindac on rectal mucosa in familial adenomatous polyposis
.
Dis Colon Rectum
1997
;
40
:
1156
9
.
59.
Richard
CS
,
Berk
T
,
Bapat
BV
,
Haber
G
,
Cohen
Z
,
Gallinger
S
.
Sulindac for periampullary polyps in FAP patients
.
Int J Colorectal Dis
1997
;
12
:
14
8
.
60.
Seow-Choen
F
,
Vijayan
V
,
Keng
V
.
Prospective randomized study of sulindac versus calcium and calciferol for upper gastrointestinal polyps in familial adenomatous polyposis
.
Br J Surg
1996
;
83
:
1763
6
.
61.
Nugent
KP
,
Farmer
KC
,
Spigelman
AD
,
Williams
CB
,
Phillips
RK
.
Randomized controlled trial of the effect of sulindac on duodenal and rectal polyposis and cell proliferation in patients with familial adenomatous polyposis
.
Br J Surg
1993
;
80
:
1618
9
.
62.
Wallace
MH
,
Forbes
A
,
Beveridge
IG
,
Spigelman
AD
,
Hewer
A
,
Venitt
S
, et al
.
Randomized, placebo-controlled trial of gastric acid-lowering therapy on duodenal polyposis and relative adduct labeling in familial adenomatous polyposis
.
Dis Colon Rectum
2001
;
44
:
1585
9
.
63.
Parc
Y
,
Desaint
B
,
Fléjou
J-F
,
Lefèvre
JH
,
Serfaty
L
,
Vienne
A
, et al
.
The effect of ursodesoxycholic acid on duodenal adenomas in familial adenomatous polyposis: a prospective randomized placebo-control trial
.
Colorectal Dis
2012
;
14
:
854
60
.
64.
Phillips
RKS
,
Wallace
MH
,
Lynch
PM
,
Hawk
E
,
Gordon
GB
,
Saunders
BP
, et al
.
A randomized, double blind, placebo controlled study of celecoxib, a selective cyclooxygenase 2 inhibitor, on duodenal polyposis in familial adenomatous polyposis
.
Gut
2002
;
50
:
857
60
.
65.
Mathers
JC
,
Elliott
F
,
Macrae
F
,
Mecklin
J-P
,
Möslein
G
,
McRonald
FE
, et al
.
Cancer prevention with resistant starch in Lynch syndrome patients in the CAPP2-randomized placebo controlled trial: planned 10-year follow-up
.
Cancer Prev Res
2022
;
15
:
623
34
.
66.
Spigelman
AD
,
Williams
CB
,
Talbot
IC
,
Domizio
P
,
Phillips
RK
.
Upper gastrointestinal cancer in patients with familial adenomatous polyposis
.
Lancet
1989
;
2
:
783
5
.
67.
Steinbach
G
,
Lynch
PM
,
Phillips
RK
,
Wallace
MH
,
Hawk
E
,
Gordon
GB
, et al
.
The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis
.
N Engl J Med
2000
;
342
:
1946
52
.
68.
West
NJ
,
Clark
SK
,
Phillips
RKS
,
Hutchinson
JM
,
Leicester
RJ
,
Belluzzi
A
, et al
.
Eicosapentaenoic acid reduces rectal polyp number and size in familial adenomatous polyposis
.
Gut
2010
;
59
:
918
25
.
69.
Lynch
PM
,
Burke
CA
,
Phillips
R
,
Morris
JS
,
Slack
R
,
Wang
X
, et al
.
An international randomized trial of celecoxib versus celecoxib plus difluoromethylornithine in patients with familial adenomatous polyposis
.
Gut
2016
;
65
:
286
95
.
70.
Burn
J
,
Bishop
DT
,
Chapman
PD
,
Elliott
F
,
Bertario
L
,
Dunlop
MG
, et al
.
A randomized placebo-controlled prevention trial of aspirin and/or resistant starch in young people with familial adenomatous polyposis
.
Cancer Prev Res
2011
;
4
:
655
65
.
71.
Ishikawa
H
,
Mutoh
M
,
Sato
Y
,
Doyama
H
,
Tajika
M
,
Tanaka
S
, et al
.
Chemoprevention with low-dose aspirin, mesalazine, or both in patients with familial adenomatous polyposis without previous colectomy (J-FAPP Study IV): a multicenter, double-blind, randomized, two-by-two factorial design trial
.
Lancet Gastroenterol Hepatol
2021
;
6
:
474
81
.