Next Article in Journal
The Potential Biological Roles of Circular RNAs in the Immune Systems of Insects to Pathogen Invasion
Previous Article in Journal
Special Issue “DNA Replication/Repair, and the DNA Damage Response in Human Disease”
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Molecular and Sociodemographic Colorectal Cancer Disparities in Latinos Living in Puerto Rico

by
Julyann Perez-Mayoral
1,†,
Maria Gonzalez-Pons
2,*,†,
Hilmaris Centeno-Girona
2,
Ingrid M. Montes-Rodríguez
2,
Marievelisse Soto-Salgado
2,
Belisa Suárez
2,
Natalia Rodríguez
3,
Giancarlo Colón
3,
Javier Sevilla
3,
Daphne Jorge
4,
Xavier Llor
5,
Rosa M. Xicola
5,
Doris H. Toro
6,
Luis Tous-López
7,
Marla Torres-Torres
7,
José S. Reyes
7,
Nicolas López-Acevedo
7,
Ajay Goel
8,
Segundo Rodríguez-Quilichini
9 and
Marcia Cruz-Correa
2,9
1
NCI Center for Cancer Genomics, Bethesda, MD 20892, USA
2
University of Puerto Rico Comprehensive Cancer Center, San Juan, PR 00936, USA
3
School of Medicine, Universidad Central del Caribe, Bayamon, PR 00956, USA
4
School of Medicine, Ponce Health Sciences University, Ponce, PR 00716, USA
5
Department of Internal Medicine and Digestive Diseases, School of Medicine, Yale University, New Haven, CT 06520, USA
6
VA Caribbean Healthcare System, San Juan, PR 00921, USA
7
Ashford Presbyterian Community Hospital, San Juan, PR 00907, USA
8
Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
9
Department of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, PR 00935, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Genes 2023, 14(4), 894; https://doi.org/10.3390/genes14040894
Submission received: 15 February 2023 / Revised: 5 April 2023 / Accepted: 7 April 2023 / Published: 11 April 2023
(This article belongs to the Section Molecular Genetics and Genomics)

Abstract

:
Background: The incidence of sporadic colorectal cancer (CRC) among individuals <50 years (early-onset CRC) has been increasing in the United States (U.S.) and Puerto Rico. CRC is currently the leading cause of cancer death among Hispanic men and women living in Puerto Rico (PRH). The objective of this study was to characterize the molecular markers and clinicopathologic features of colorectal tumors from PRH to better understand the molecular pathways leading to CRC in this Hispanic subpopulation. Methods: Microsatellite instability (MSI), CpG island methylator phenotype (CIMP), and KRAS and BRAF mutation status were analyzed. Sociodemographic and clinicopathological characteristics were evaluated using Chi-squared and Fisher’s exact tests. Results: Of the 718 tumors analyzed, 34.2% (n = 245) were early-onset CRC, and 51.7% were males. Among the tumors with molecular data available (n = 192), 3.2% had MSI, 9.7% had BRAF, and 31.9% had KRAS mutations. The most common KRAS mutations observed were G12D (26.6%) and G13D (20.0%); G12C was present in 4.4% of tumors. A higher percentage of Amerindian admixture was significantly associated with early-onset CRC. Conclusions: The differences observed in the prevalence of the molecular markers among PRH tumors compared to other racial/ethnic groups suggest a distinct molecular carcinogenic pathway among Hispanics. Additional studies are warranted.

1. Introduction

Colorectal cancer (CRC) is the second leading cause of cancer death among men and women in the United States (U.S.) [1]. Disparities in CRC incidence and survival have been well documented among racial/ethnic groups in the mainland U.S. [2,3]. Although CRC incidence and mortality rates are lower among U.S. mainland Hispanics than in other racial/ethnic groups, aggregating heterogeneous populations (e.g., Hispanics) may mask the significant variability in CRC incidence and mortality within subgroups [4]. For example, among the Hispanic subgroups living on the U.S. mainland, Cubans and Puerto Ricans have disproportionately higher CRC incidence and mortality rates [5,6]. In Puerto Rico, CRC is the leading cause of cancer death in men and women [7].
During the past 30 years, the incidence of sporadic, non-familial CRC among individuals <50 years (early-onset CRC) has been increasing at an alarming rate in the U.S. and is expected to increase by >140% by 2030 [8,9,10]. In the U.S., more than 11% of CRC cases and 6% of deaths were reported due to early-onset CRC during 2012–2017 [11]. During the same period in Puerto Rico, more than 9% of the CRC cases and approximately 6% of CRC deaths corresponded to patients <50 years old [7]. Despite the attention the ascending CRC incidence rates among young individuals have garnered, the molecular events that lead to the development of early-onset CRC remain poorly understood.
Sporadic CRC is a heterogeneous disease that arises from the gradual accumulation of genetic and epigenetic alterations, some of which can be used as molecular markers to inform medical treatment decisions (i.e., MSI, BRAF or KRAS mutations) [12]. The CpG island methylator phenotype (CIMP) is another molecular feature associated with CRC [13]; however, the role of the CIMP in colorectal carcinogenesis is still not clearly understood. Based on the combination of these molecular markers (MSI, CIMP, and BRAF and KRAS mutations), colorectal tumors can be classified according to three carcinogenic pathways [14,15]. The traditional pathway, which leads to approximately 50–70% of all CRC cases, is characterized by chromosomal instability and APC and KRAS mutations resulting in CIMP-negative and MSI-low or microsatellite stable (MSS) tumors with a predominant distal location. Up to 30% of colorectal tumors are estimated to arise from the alternate carcinogenesis pathway, where KRAS or APC mutations precede the development of CIMP-low, MSI-low, or MSS tumors. In the serrated pathway, a BRAF mutation leads to MSI-high, MSI-low, or MSS, CIMP-high tumors that comprise 10–20% of all CRC cases [16].
There is limited information regarding the pathways that lead to CRC among Hispanics, as most studies have focused on non-Hispanic Whites and non-Hispanic Blacks [17,18]. This study describes the molecular and clinicopathological characteristics of CRC tumors in Hispanics living in Puerto Rico (PRH), a Hispanic subpopulation with a disproportionate CRC burden. A better understanding of the molecular events leading to colorectal carcinogenesis in Hispanic subpopulations, such as PRH, is necessary to develop tailored prevention and/or treatment strategies to promote health equity and reduce CRC mortality.

2. Materials and Methods

2.1. Patient Recruitment, Sociodemographic, and Clinicopathological Characteristics

A total of 718 sporadic, non-familial colorectal tumors from PRH were recruited through the Puerto Rico Familial Colorectal Cancer Registry. This island-wide, population-based registry recruits and collects biospecimens from individuals with gastrointestinal neoplasia and healthy controls. Fresh frozen tumor tissues were collected during tumor resection and stored at −150 °C for future analysis. According to the U.S. Census, “Hispanic or Latino” is defined as a person of Cuban, Mexican, Puerto Rican, South or Central American, or other Spanish culture or origin regardless of race. All participants in the Puerto Rico Familial Colorectal Cancer Registry complete a questionnaire (in Spanish) modeled from the one used in the Collaborative Family Registries for Colorectal Cancer. This questionnaire collects information on which country the participant was born in as well as the country where their father and mother were born, allowing us to classify subjects as Hispanic.
For this study, exclusion criteria included having a diagnosis of any hereditary genetic syndrome (i.e., familial adenomatous polyposis, Lynch syndrome) or inflammatory bowel disease. Only adenocarcinomas confirmed by pathology were included. The sociodemographic data analyzed included: age at recruitment (<50 vs. ≥50 years of age), gender (female vs. male), marital status (single/divorced/widowed vs. living together/married), educational level (<high school diploma or equivalent vs. ≥high school diploma or equivalent), type of health insurance (private vs. public vs. Medicare/Medicare Advantage), current drinker (yes vs. no) and current smoker (yes vs. no). The clinicopathological characteristics examined were body mass index (BMI kg/m2; <25, underweight/normal vs. ≥25, overweight/obese), family history of CRC (yes vs. no), tumor location (proximal vs. distal vs. unknown), tumor differentiation (high vs. moderate or low), and tumor stage (I/II vs. III/IV).

2.2. Genomic DNA Extraction

Genomic DNA colorectal tissue was extracted using a Gentra Puregene Tissue Kit (Qiagen, Germantown, MD, U.S.), following the manufacturer’s protocol. DNA concentrations were quantified using a Nanodrop (Thermo Scientific, Waltham, MA, U.S.). Samples with 260/280 ratio of ~1.8 were subsequently subjected to PCR analysis using β-actin primers to assay DNA integrity. Samples from which β-actin could not be amplified were excluded from the study. A representative agarose gel electrophoresis assay showing amplification of the expected 586 bp amplicon is shown in Supplementary Figure S1.

2.3. MSI, KRAS, and BRAF Status

Microsatellite instability (MSI) status was obtained by analyzing tumor samples and corresponding normal mucosa as previously described [19,20]. The MSI testing panel used included the following markers: BAT-25, BAT-26, BAT-40, D2S123, D5SS346, D175250, and TGFBR2. These markers included the reference panel of five MSI markers recommended by the National Cancer Institute for colorectal tumors; loci were scored according to published guidelines [21]. The mutation status of KRAS and BRAF was obtained from pathology reports or by performing a custom real-time PCR assay using a qBiomarker Somatic Mutation PCR Array (cat # 337021) following the manufacturer’s instructions (Qiagen, Germantown, MD, U.S.). This somatic mutation PCR array was a custom TaqMan-based mutation panel designed specifically to a restricted number of mutations in KRAS and BRAF, including: 1 BRAF (COSMIC ID: COSM476 or V600E (cat # SMPH001828A) and 11 KRAS somatic mutations (COSMIC ID: COSM516 (cat #SMPH007535A), COSM517 (cat #SMPH007533A), COSM520 (cat #SMPH007537A), COSM521 (cat #SMPH007531A), COSM522 (cat #SMPH007536A), COSM527 (cat #SMPH007541A), COSM531 (cat #SMPH007589A), COSM532 (cat #SMPH007538A), COSM553 (cat #SMPH007544A), COSM554 (cat #SMPH007540A), COSM555 (cat #SMPH007546A)). The master mix consisted of a 2X qBiomarker Probe Mix, genomic DNA (50 ng/μL), and RNase-free water, in a 25 μL total reaction volume. The real-time PCR was carried out in a StepOnePlus™ Real-Time PCR System (Applied Biosystems, Waltham, MA, U.S.) with the following cycling parameters: 95 °C for 10 min, 40 cycles of 95 °C for 15 s, 60 °C for 60 s. Supplementary Figure S2 shows a quantification plot of one sample for all the analyzed mutations.

2.4. CpG Island Methylation Phenotype (CIMP) Analysis

Bisulfite conversion was performed on 300 ng of genomic tumor DNA using a methylSEQr Kit (Applied Biosystems, Waltham, MA, U.S.) following the manufacturer’s protocol. A total of 2 μL of the bisulfite-modified DNA was used for the subsequent methylation-specific PCR (MSP) analysis performed using primers specific for the eight genes in the CIMP panel: CAGNA1G, CRABP1, NEUROG1, IGF2, RUNX3, SOCS1, CDKN2 and MLH1 [22,23]. The conditions for the MSP were as follows: 95 °C for 10 min, followed by 40 cycles of 94 °C for 30 s, the annealing temperature of the primer set for 40 s, 72 °C for 40 s, with a final extension of 72 °C for 10 min. Annealing temperatures and primer sequences for each gene in the CIMP panel are described in Supplementary Table S1. After MSP, PCR products were visualized in 2% agarose gel and documented using the Gel Doc 1000 system with molecular analysis software (Bio-Rad, Hercules, CA, U.S.). Tumors were classified according to CIMP status as follows: CIMP-Zero (0 methylated genes), CIMP-Low (one to five methylated genes), and CIMP-High (six to eight methylated genes) [23].

2.5. Ancestry Informative Markers (AIMS) Panel Genotyping

PBL genomic DNA was used to genotype 105 AIMs panels using the Sequenom MassArray iPLEX platform (Sequenom, San Diego, CA, U.S.) as described in Perez-Mayoral et al. 2019. This AIMs panel consists of SNP markers that inform European, African, and Amerindian ancestry and has been validated for estimating continental ancestry information in admixed Latino populations, including Puerto Ricans. Sequenom TYPER software (Sequenom, San Diego, CA, U.S.) was used to make genotype SNP calls. STRUCTURE v2.3 software (Stanford, CA, U.S.) was used to calculate individual ancestry estimates for each participant using a model-based clustering method.

2.6. Integrated Pathways

Tumors were classified according to their molecular characteristics into the traditional pathway (MSS, CIMP-negative, and/or BRAF and KRAS-wild type), serrated pathway (BRAF-mutated, CIMP-positive, and any MSI or MSS), alternative pathway (MSS, CIMP-L, and KRAS-mutated), or other pathway [15,24,25]. Cases with unavailable data for the analyzed markers were classified into the traditional, serrated, or alternate pathways if two or more markers were available. Otherwise, cases were classified as other pathway.

2.7. Statistical Analysis

Frequency distributions and percentages were generated to describe the sociodemographic and clinicopathological characteristics of the sample. The differences between the variables were evaluated using Fisher’s exact test or the Chi-squared test. All statistical analyses were performed using STATA 16.0 (StataCorp LLC, College Station, TX, U.S.). Comparisons between PRH and other racial/ethnic groups were tested using a two-sample proportion test (prtesti). A p-value < 0.05 was used to determine statistical significance. This value indicates the presence of significant differences among the compared groups. However, given the cross-sectional nature of the study, inferences on causality are not possible.

3. Results

3.1. Description of the Study Population

A total of 718 subjects with sporadic CRC were recruited during 2007–2017. Among the cases included in the study, 34.2% of the subjects were diagnosed with CRC before 50 years of age (early-onset CRC), 51.7% were male, 60.7% had ≥12 years of education, and most had European admixture (Table 1). Most of the tumors evaluated were from CRC patients with no family history of CRC (72.3%), were in the distal colon (72.6%), and were diagnosed at early stages (stage I or II; 71.9%).

3.2. Description of the CRC Tumors with Molecular Markers

Of the 718 tumors, we had molecular marker data for 192 cases. Molecular testing of CRC tumors showed that BRAF- and KRAS-mutation status, MSS, and CIMP overall age distribution were similar among those diagnosed with early-onset CRC (< 50 years) and later-onset CRC (50 years) (Table 2). A higher frequency of wild-type KRAS was observed among later-onset CRC cases (72.9% p = 0.077); these results were marginally significant. A slightly higher frequency of MSI-high (5.6%) and CIMP-low (92.3%) was observed among early-onset CRC cases. MSI was only detected in 6 out of 186 cases, and 1 out of 111 cases had the CIMP-high phenotype.

3.3. BRAF/KRAS Mutation Status

The KRAS mutation spectrum was evaluated in 30 CRC tumors (Table 3). The most common somatic mutations found were G12D (26.6%) and G13D (20.0%). Both of these changes were predicted to be pathogenic with a pathogenicity score of 0.98 based on the Functional Analysis through Hidden Markov Models (v2.3) in silico model [26].

3.4. CRC Pathways

Of the 186 CRC tumors with two or more molecular markers, the majority (49.5%) were classified into the “other pathways” category (Table 4). When comparing the CRC pathways according to ancestry, a significantly higher number of cases had a higher percentage of Amerindian admixture in the traditional and other pathways (p = 0.029). A higher number of individuals with tumors from the alternate pathway reported < high school diploma or equivalent education (54.3%, p < 0.05).

3.5. Characteristics According to CRC Diagnostic Age

Considering the increasing trend in early-onset CRC incidence, we compared the sociodemographic and clinicopathological characteristics according to age at diagnosis (n = 717; Table 5). A higher percentage of females was diagnosed with early-onset CRC (58.4%) compared to later-onset CRC (43.2%; p < 0.05). A significantly lower rate of European admixture and higher Amerindian admixture was detected in individuals younger than 50 years diagnosed with CRC. Compared to individuals with later-onset CRC, those younger than 50 years of age when diagnosed with CRC had higher educational levels, private insurance, and no family history of CRC (p < 0.05).

3.6. Comparison of BRAF, KRAS, Microsatellite Instability and CIMP Status among Different Populations

Differences in the prevalence of the studied markers and the gender variable were found when comparing PRH with other global populations (Table 6). Compared to PRH, the Spanish and U.S. populations had a higher proportion of males by 7.3% and a lower proportion of females by 7.3% and 6.7%, respectively, across both genders. PRH had a higher prevalence of BRAF mutations (9.7%) than what was reported among the Spanish (6.2%; p < 0.05), Chinese (0.7%; p < 0.001), and two U.S. cohort studies, the Nurses’ Health Study (NHS) and the Health Professionals Follow-up Study (HPFS) (14.6%; p > 0.05) [27,28,29]. The prevalence of KRAS mutations in PRH (31.9%) was higher than in the Chinese group (24.9%) but lower than in the Spanish (36.9%) and U.S. cohorts (35.9%), although these differences were not statistically significant. CIMP-H status was markedly lower in PRH (0.9%) compared to the Spanish, Chinese, and U.S. populations (27.4%, 10%, and 17.5%, respectively). In addition, the MSI-H status of PRH was lower than that of the Chinese population (6.1%; p > 0.05) and significantly lower than that of the U.S. cohort (15.4%; p < 0.001) [27,28,29].

4. Discussion

Although CRC incidence and mortality rates are lower among U.S. mainland Hispanics than in other racial/ethnic groups, CRC continues to be one of the leading causes of cancer mortality among Hispanics [30]. Racial/ethnic early-onset CRC disparities have also been reported, with Hispanics having the most marked increases in early-onset CRC incidence annually [31,32]; however, the molecular events leading to the development of early-onset CRC and the racial/ethnic disparities observed remain poorly understood. One of the limitations in most studies that include Hispanics is that individuals from different countries of origin are usually classified together as “Hispanics”, which may mask the significant variability in CRC incidence and mortality within subgroups [4]. In this study, we report the molecular characterization of CRC tumors from Puerto Rican Hispanics, the second-largest Hispanic subpopulation in the mainland U.S., for the first time.
Analysis of the molecular biomarker data available for tumors in our PRH showed that the frequency of BRAF mutations, MSI, CIMP status, and KRAS mutations was different from what was reported for tumors from individuals from other racial/ethnic groups. The frequency of BRAF mutations found in PRH tumors overall (9.7%) is noticeably lower than that in non-Hispanic Blacks (56%) and non-Hispanic Whites (11.5–44%) [33,34,35,36]. Previous reports have also established an association between BRAF mutations and MSI status [35,37,38]; this association was not observed in our cohort. Colorectal tumors in the proximal colon were reported to be more likely to have BRAF V600E mutations and the CIMP-high phenotype than tumors in the distal colon [39,40]. The majority (72.6%) of the tumors included in the study were in the distal colon, which could in part explain the low rates of BRAF mutations and CIMP-high tumors. Overall, KRAS mutation frequencies in our PRH cohort (31.9%) are lower than those reported in other racial/ethnic groups, including non-Hispanic Blacks (59%) and non-Hispanic Whites (37–41%) [33,34,35,41]. A previous study in a separate cohort of 501 PRH reported a slightly higher but comparable prevalence of KRAS mutations (39%) [42]. The percentage of tumors with MSI in our cohort was markedly lower (4.3%) than what has been reported in African Americans (14–19%) and non-Hispanic Whites (9–13%) [33,37,43,44]. A very low number of tumors in our cohort had CIMP, with only one case having the CIMP-high phenotype. This result is markedly lower than the reported prevalence of CIMP-high among non-Hispanic Whites (13%), non-Hispanic Blacks (4.5%), and Hispanics (12.3%) [45]. We compared the prevalence of tumor markers from PRH with that reported for the nationwide and multicenter Spanish EPICOLON I and EPICOLON II projects, a Chinese population study, and the U.S. cohort studies, NHS and HPFS [27,28,29]. PRH has a higher prevalence of BRAF mutations than the Spanish and Chinese populations, but a lower prevalence than the U.S. study cohort. The prevalence of KRAS mutations ranges from 24.9% to 35.9% across populations, with the U.S. having the highest prevalence and the Chinese having the lowest. When compared, the Spanish cohort had the highest CIMP-H status, while the PRH cohort had the lowest. On the other hand, the U.S. had the highest percentage of MIS-H cases, while PRH had the lowest. Importantly, both MSI-L and MSI-H were reported as MSI by the Spanish group. The differences in the frequencies of the tumor biomarkers in our study population and other racial/ethnic groups in the mainland U.S. and other global populations can be attributed to various factors, including genetic (e.g., population-specific variations) and environmental exposures and diet, among others [46,47,48]. Although Hispanics share a common language and history, according to the country of origin, Hispanic subpopulations have different diets, exposures, and degrees of European, African, and Native American genetic admixture, which may explain the differences in colorectal tumor biomarkers in our population [49,50].
Approximately 90% of the somatic mutations found in KRAS in colorectal cancer tumors are in codons 12, 13, and 61 [51,52]. The most common mutations in KRAS observed in our cohort were G12D and G13D; these two mutations were found to be pathogenic according to the FATHMM in silico model [26]. G12V and G12C, some of the most commonly reported G12 KRAS mutations [53], represented a low percentage of the KRAS mutations detected in our population. KRAS mutation status is currently used as a prognostic factor for anti-EGFR therapies [54]. Recent studies suggest that individuals with mutations in codon 13 could derive benefits from anti-EGFR therapy [55,56], but individuals with somatic mutations in KRAS codon 12 have been reported to have worse overall survival than individuals with other KRAS somatic mutations [57]. Although the KRAS mutation rates in our PRH cohort were markedly lower than what has been reported in non-Hispanic Blacks, the higher frequency of KRAS mutations in codon 12 could be a contributing factor to the comparable and significantly higher relative risk of CRC death reported for PRH and non-Hispanic Blacks compared to non-Hispanic Whites [2].
The genetic and epigenetic alterations that lead to colorectal carcinogenesis can be grouped into three major pathways: traditional pathway (MSS, CIMP-negative, and/or wild type BRAF and KRAS), serrated pathway (MSI or MSS, CIMP-positive, BRAF mutation), and alternate pathway (MSS, CIMP-low, KRAS mutation). Most of the tumors in our PRH cohort were classified into the other pathway category, supporting the theory that distinct population-specific variation, genetic/epigenetic aberrations, and environmental factors may contribute to the carcinogenic process. A significantly higher number of individuals with tumors in the traditional and other pathway had a higher percentage of Amerindian admixture. Information on the prevalence of CRC molecular pathways according to race/ethnicity and genetic admixture is lacking, and warrants investigation. Our group previously reported that PRH with higher levels of African ancestry were three times more likely to develop colorectal tumors located in the rectum [58]. These findings show that genetic ancestry may have a role in the molecular development of CRC and further studies are needed to fully elucidate its contribution to colorectal carcinogenesis in diverse populations in order to develop tailored screening and treatment strategies to improve CRC outcomes.
Among our population, significant differences in gender, family history, and genetic admixture were observed when comparing early-onset versus later-onset CRC (>50 years at diagnosis). A higher number of women was diagnosed with early-onset CRC compared to men. This is in contrast to a large nationwide study reporting that men have 16% higher incidence rates of early-onset CRC [59]. As most early-onset CRC cases in our study were diagnosed at early stages, a possible reason that may in part explain this disparity is that women have been reported to seek more healthcare compared to men [60]. However, environmental exposures, in utero exposures, or epigenomic factors, among others may contribute to the observed gender-specific difference among this early onset cohort. The significantly higher number of early-onset CRC cases with family history of CRC compared to later-onset cases observed in our population is consistent with previous studies showing that family history of CRC is associated with early-onset CRC [61] and that some common CRC risk variants are more strongly associated with early-onset CRC than later-onset CRC [62]. Significantly lower levels of European admixture and higher levels of Amerindian admixture were observed among individuals with early-onset CRC. Hispanics vary in their percentages of admixture of ancestral population and in the fact that they are the racial/ethnic subgroup in the U.S. with the highest increase in annual incidence rates from 2013–2018 [59]. Thus, larger studies evaluating the association between genetic admixture and early-onset CRC are needed to determine if this is a factor that could be used to identify individuals at higher risk of developing CRC at an early age and to develop tailored screening guidelines according to admixture.
The strength of this study is that it characterizes the sociodemographic and clinical characteristics, as well as the molecular markers, from CRC cases diagnosed from PRH, a Hispanic subpopulation with a high CRC cancer burden. All subjects were recruited through PURIFICAR, a population-based registry that receives direct referrals from physicians and surgeons across the island. However, this could create a selection bias, and the data presented may not be representative of the PRH population. Another limitation is that not all the CRC cases recruited had all the molecular biomarkers examined in this study performed as part of their clinical workup. We were unable to perform molecular testing on tumors if subjects were recruited into the registry after surgery and we did not have access to the tumor tissue. Molecular testing was performed on all the tumor tissue we had available at the time of the study. The size of various subgroups, such as CRC tumors with MSI-high, limited the statistical analysis performed for this study, thus warranting a study with a larger sample size to be able to perform robust subgroup analysis, as well as to evaluate lifestyle CRC risk factors among PRH. In addition, future studies including a more comprehensive array of ”omic” data are warranted for CRC tumors among PRH in order to classify tumors into the CRC consensus molecular subtypes [25] and for the development of personalized treatment strategies to improve outcomes.
To the best of our knowledge, this is the first study to characterize tumors from PRH patients using MSI, CIMP, and KRAS and BRAF mutation status. The observation that the prevalence of these molecular markers is markedly different from what has been reported in other racial/ethnic groups suggests distinct pathways for CRC carcinogenesis in Hispanic populations. Moreover, the lower percentage of European admixture and higher Amerindian admixture detected in PRH with early-onset CRC supports the need for additional studies with larger sample sizes to examine ancestry, genetics, epigenetics, and lifestyle to fully understand and characterize the factors contributing to the development of early- and later-onset CRC in Hispanic subpopulations.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14040894/s1, Table S1: Primer Information for CIMP Panel Genes, Figure S1: DNA integrity assessed by β-actin PCR analysis; Figure S2: TaqMan-based mutation panel analysis.

Author Contributions

The first two authors contributed equally to this work; M.G.-P. is the corresponding author. M.G.-P., J.P.-M., B.S., N.R., G.C., J.S., D.J., X.L., R.M.X. and A.G. conducted data acquisition; D.H.T., L.T.-L., M.T.-T., J.S.R., N.L.-A. and S.R.-Q. helped recruit the subjects and collect tissues; M.G.-P., J.P.-M. and M.C.-C. conceptualized the study; H.C.-G. and M.S.-S. performed statistical analysis; M.G.-P., J.P.-M., I.M.M.-R. and H.C.-G. wrote the manuscript; M.C.-C. critically reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by the National Cancer Institute (award numbers K22CA115913, R21CA167220, R03CA130034, and U54CA096297), the RCMI Center for Collaborative Research in Health Disparities (NIMHD award number G12MD007600), and the Hispanic Alliance for Clinical and Translational Research (NIGMS award number U54GM133807). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. In addition, we acknowledge that this manuscript is also a result of work supported with resources and the use of facilities at the VA Caribbean Healthcare System. The contents of this publication do not necessarily represent the views of the VA Caribbean Healthcare System, the Department of Veterans Affairs, or the United States Government.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the University of Puerto Rico Medical Sciences Campus Institutional Review Board (#A2210207).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data are available through a controlled access repository at https://crcweb.rcm.upr.edu/redcap/. Researchers are required to submit a web-based application to request access to the data. Once the request is approved, access will be granted.

Acknowledgments

The authors would like to thank the participants of the Puerto Rico Familial Colorectal Cancer Registry for their contribution.

Conflicts of Interest

MCC reports research support from the following pharmaceutical companies: Abbvie, BMS, Regeneron, QED, SeaGen, Janssen, TAIHO, BeiGene, MIRATI, Merck, Gilead, Pfizer, Natera, Genentech, Elli Lilly, and Apollomics. The content of this manuscript is not related to the research support from the pharmaceutical industry.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Gonzalez-Pons, M.; Torres, M.; Perez, J.; Velez, A.; Betancourt, J.P.; Marcano, L.; Soto-Salgado, M.; Cruz-Correa, M. Colorectal Cancer Survival Disparities among Puerto Rican Hispanics: A Comparison to Racial/Ethnic Groups in the United States. Cancer Clin. Oncol. 2016, 5, 29–37. [Google Scholar] [CrossRef] [PubMed]
  3. Zavala, V.A.; Bracci, P.M.; Carethers, J.M.; Carvajal-Carmona, L.; Coggins, N.B.; Cruz-Correa, M.R.; Davis, M.; de Smith, A.J.; Dutil, J.; Figueiredo, J.C.; et al. Cancer health disparities in racial/ethnic minorities in the United States. Br. J. Cancer 2020, 124, 315–332. [Google Scholar] [CrossRef] [PubMed]
  4. Siegel, R.L.; Miller, K.D.; Sauer, A.G.; Fedewa, S.A.; Butterly, L.F.; Anderson, J.C.; Cercek, A.; Smith, R.A.; Jemal, A. Colorectal cancer statistics. CA Cancer J. Clin. 2020, 70, 145–164. [Google Scholar] [CrossRef] [Green Version]
  5. Pinheiro, P.S.; Callahan, K.E.; Siegel, R.L.; Jin, H.; Morris, C.R.; Trapido, E.J.; Gomez, S.L. Cancer Mortality in Hispanic Ethnic Groups. Cancer Epidemiol. Biomark. Prev. 2017, 26, 376–382. [Google Scholar] [CrossRef] [Green Version]
  6. Miller, K.D.; Sauer, A.G.; Ortiz, A.P.; Fedewa, S.A.; Pinheiro, P.S.; Tortolero-Luna, G.; Martinez-Tyson, D.; Jemal, A.; Siegel, R.L. Cancer Statistics for Hispanics/Latinos. CA A Cancer J. Clin. 2018, 68, 425–445. [Google Scholar] [CrossRef] [Green Version]
  7. Torres-Cintrón, C.R.; Alvarado-Ortiz, M.; Román-Ruiz, Y.; Ortiz-Ortiz, K.J.; Zavala-Zegarra, D.; Tortolero-Luna, G. Cancer in Puerto Rico, Puerto Rico Central Cancer Registry. San Juan, Puerto Rico. 2012.
  8. Bailey, C.E.; Hu, C.-Y.; You, Y.N.; Bednarski, B.K.; Rodriguez-Bigas, M.A.; Skibber, J.M.; Cantor, S.B.; Chang, G.J. Increasing Disparities in the Age-Related Incidences of Colon and Rectal Cancers in the United States. JAMA Surg. 2015, 150, 17–22. [Google Scholar] [CrossRef] [Green Version]
  9. Mauri, G.; Sartore-Bianchi, A.; Russo, A.G.; Marsoni, S.; Bardelli, A.; Siena, S. Early-onset colorectal cancer in young individuals. Mol. Oncol. 2018, 13, 109–131. [Google Scholar] [CrossRef] [Green Version]
  10. Patel, S.G.; Ahnen, D.J. Colorectal Cancer in the Young. Curr. Gastroenterol. Rep. 2018, 20, 15. [Google Scholar] [CrossRef]
  11. Caporaso, J.G.; Lauber, C.L.; Walters, W.A.; Berg-Lyons, D.; Lozupone, C.A.; Turnbaugh, P.J.; Fierer, N.; Knight, R. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc. Natl. Acad. Sci. USA 2011, 108 (Supp. S1), 4516–4522. [Google Scholar] [CrossRef] [Green Version]
  12. Jass, J.R. Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathology 2006, 50, 113–130. [Google Scholar] [CrossRef] [PubMed]
  13. Issa, J.-P. CpG island methylator phenotype in cancer. Nat. Rev. Cancer 2004, 4, 988–993. [Google Scholar] [CrossRef] [PubMed]
  14. De Palma, F.D.E.; D’Argenio, V.; Pol, J.; Kroemer, G.; Maiuri, M.C.; Salvatore, F. The Molecular Hallmarks of the Serrated Pathway in Colorectal Cancer. Cancers 2019, 11, 1017. [Google Scholar] [CrossRef] [Green Version]
  15. Leggett, B.; Whitehall, V. Role of the Serrated Pathway in Colorectal Cancer Pathogenesis. Gastroenterology 2010, 138, 2088–2100. [Google Scholar] [CrossRef] [PubMed]
  16. Yamagishi, H.; Kuroda, H.; Imai, Y.; Hiraishi, H. Molecular pathogenesis of sporadic colorectal cancers. Chin. J. Cancer 2016, 35, 4. [Google Scholar] [CrossRef] [Green Version]
  17. Guinney, J.; Dienstmann, R.; Wang, X.; De Reyniès, A.; Schlicker, A.; Soneson, C.; Marisa, L.; Roepman, P.; Nyamundanda, G.; Angelino, P.; et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 2015, 21, 1350–1356. [Google Scholar] [CrossRef]
  18. Inamura, K. Colorectal Cancers: An Update on Their Molecular Pathology. Cancers 2018, 10, 26. [Google Scholar] [CrossRef] [Green Version]
  19. Goel, A.; Arnold, C.N.; Niedzwiecki, D.; Chang, D.K.; Ricciardiello, L.; Carethers, J.M.; Dowell, J.M.; Wasserman, L.; Compton, C.; Mayer, R.J.; et al. Characterization of sporadic colon cancer by patterns of genomic instability. Cancer Res 2003, 63, 1608–1614. [Google Scholar]
  20. Xicola, R.M.; Llor, X.; Pons, E.; Castells, A.; Alenda, C.; Piñol, V.; Andreu, M.; Castellvi-Bel, S.; Payá, A.; Jover, R.; et al. Performance of Different Microsatellite Marker Panels for Detection of Mismatch Repair–Deficient Colorectal Tumors. Gynecol. Oncol. 2007, 99, 244–252. [Google Scholar] [CrossRef]
  21. Boland, C.R.; Thibodeau, S.N.; Hamilton, S.R.; Sidransky, D.; Eshleman, J.R.; Burt, R.W.; Meltzer, S.J.; Rodriguez-Bigas, M.A.; Fodde, R.; Ranzani, G.N.; et al. A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: Development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 1998, 58, 5248–5257. [Google Scholar]
  22. Weisenberger, D.J.; Siegmund, K.D.; Campan, M.; Young, J.; Long, T.I.; Faasse, M.A.; Kang, G.H.; Widschwendter, M.; Weener, D.; Buchanan, D.; et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat. Genet. 2006, 38, 787–793. [Google Scholar] [CrossRef] [PubMed]
  23. Ogino, S.; Kawasaki, T.; Kirkner, G.J.; Kraft, P.; Loda, M.; Fuchs, C.S. Evaluation of Markers for CpG Island Methylator Phenotype (CIMP) in Colorectal Cancer by a Large Population-Based Sample. J. Mol. Diagn. 2007, 9, 305–314. [Google Scholar] [CrossRef] [Green Version]
  24. Samadder, N.; Vierkant, R.A.; Tillmans, L.S.; Wang, A.H.; Weisenberger, D.J.; Laird, P.W.; Lynch, C.F.; Anderson, K.E.; French, A.J.; Haile, R.W.; et al. Associations Between Colorectal Cancer Molecular Markers and Pathways With Clinicopathologic Features in Older Women. Gastroenterology 2013, 145, 348–356. [Google Scholar] [CrossRef] [Green Version]
  25. Phipps, A.I.; Alwers, E.; Harrison, T.; Banbury, B.; Brenner, H.; Campbell, P.T.; Chang-Claude, J.; Buchanan, D.; Chan, A.T.; Farris, A.B.; et al. Association Between Molecular Subtypes of Colorectal Tumors and Patient Survival, Based on Pooled Analysis of 7 International Studies. Gastroenterology 2020, 158, 2158–2168. [Google Scholar] [CrossRef] [PubMed]
  26. Shihab, H.A.; Gough, J.; Cooper, D.N.; Stenson, P.D.; Barker, G.L.A.; Edwards, K.J.; Day, I.N.M.; Gaunt, T.R. Predicting the Functional, Molecular, and Phenotypic Consequences of Amino Acid Substitutions using Hidden Markov Models. Hum. Mutat. 2013, 34, 57–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Lochhead, P.; Kuchiba, A.; Imamura, Y.; Liao, X.; Yamauchi, M.; Nishihara, R.; Qian, Z.R.; Morikawa, T.; Shen, J.; Meyerhardt, J.A.; et al. Microsatellite Instability and BRAF Mutation Testing in Colorectal Cancer Prognostication. Gynecol. Oncol. 2013, 105, 1151–1156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Murcia, O.; Juárez, M.; Rodríguez-Soler, M.; Hernández-Illán, E.; Giner-Calabuig, M.; Alustiza, M.; Egoavil, C.; Castillejo, A.; Alenda, C.; Barberá, V.; et al. Colorectal cancer molecular classification using BRAF, KRAS, microsatellite instability and CIMP status: Prognostic implications and response to chemotherapy. PLoS ONE 2018, 13, e0203051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Liu, J.; Tang, L.; Yi, J.; Li, G.; Lu, Y.; Xu, Y.; Zhao, S.; Mao, R.; Li, X.; Ren, L.; et al. Unique characteristics of CpG island methylator phenotype (CIMP) in a Chinese population with colorectal cancer. BMC Gastroenterol. 2019, 19, 173. [Google Scholar] [CrossRef] [Green Version]
  30. Siegel, R.L.; Fedewa, S.A.; Miller, K.D.; Goding-Sauer, A.; Pinheiro, P.S.; Martinez-Tyson, D.; Jemal, A. Cancer statistics for Hispanics/Latinos. CA A Cancer J. Clin. 2015, 65, 457–480. [Google Scholar] [CrossRef] [Green Version]
  31. Gabriel, E.; Ostapoff, K.; Attwood, K.; Al-Sukhni, E.; Boland, P.; Nurkin, S. Disparities in the Age-Related Rates of Colorectal Cancer in the United States. Am. Surg. 2017, 83, 640–647. [Google Scholar] [CrossRef]
  32. Jafri, N.S.; Gould, M.; El-Serag, H.B.; Duan, Z.; Davila, J.A. Incidence and Survival of Colorectal Cancer Among Hispanics in the United States: A Population-Based Study. Dig. Dis. Sci. 2012, 58, 2052–2060. [Google Scholar] [CrossRef] [PubMed]
  33. Sylvester, B.E.; Huo, D.; Khramtsov, A.; Zhang, J.; Smalling, R.V.; Olugbile, S.; Polite, B.N.; Olopade, O.I. Molecular Analysis of Colorectal Tumors within a Diverse Patient Cohort at a Single Institution. Clin. Cancer Res. 2012, 18, 350–359. [Google Scholar] [CrossRef] [Green Version]
  34. Yuen, S.T.; Davies, H.; Chan, T.L.; Ho, J.W.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Tsui, W.W.; Chan, A.S.; et al. Similarity of the phenotypic patterns associated with BRAF and KRAS mutations in colo-rectal neoplasia. Cancer Res. 2002, 62, 6451–6455. [Google Scholar] [PubMed]
  35. Fransén, K.; Klintenäs, M.; Österström, A.; Dimberg, J.; Monstein, H.-J.; Söderkvist, P. Mutation analysis of the BRAF, ARAF and RAF-1 genes in human colorectal adenocarcinomas. Carcinogenesis 2003, 25, 527–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Beg, S.; Siraj, A.K.; Prabhakaran, S.; Bu, R.; Al-Rasheed, M.; Sultana, M.; Qadri, Z.; Al-Assiri, M.; Sairafi, R.; Al-Dayel, F.; et al. Molecular markers and pathway analysis of colorectal carcinoma in the Middle East. Cancer 2015, 121, 3799–3808. [Google Scholar] [CrossRef] [Green Version]
  37. Kumar, K.; Brim, H.; Giardiello, F.; Smoot, D.T.; Nouraie, M.; Lee, E.L.; Ashktorab, H. Distinct BRAF (V600E) and KRAS Mutations in High Microsatellite Instability Sporadic Colorectal Cancer in African Americans. Clin. Cancer Res. 2009, 15, 1155–1161. [Google Scholar] [CrossRef] [Green Version]
  38. Samowitz, W.S.; Sweeney, C.; Herrick, J.; Albertsen, H.; Levin, T.R.; Murtaugh, M.A.; Wolff, R.K.; Slattery, M.L. Poor Survival Associated with the BRAF V600E Mutation in Microsatellite-Stable Colon Cancers. Cancer Res. 2005, 65, 6063–6069. [Google Scholar] [CrossRef] [Green Version]
  39. Wang, J.; Shen, J.; Huang, C.; Cao, M.; Shen, L. Clinicopathological Significance of BRAFV600E Mutation in Colorectal Cancer: An Updated Meta-Analysis. J. Cancer 2019, 10, 2332–2341. [Google Scholar] [CrossRef] [Green Version]
  40. Ang, P.W.; Loh, M.; Liem, N.; Lim, P.L.; Grieu, F.; Vaithilingam, A.; Platell, C.; Yong, W.P.; Iacopetta, B.; Soong, R. Comprehensive profiling of DNA methylation in colorectal cancer reveals subgroups with distinct clinicopathological and molecular features. BMC Cancer 2010, 10, 227–228. [Google Scholar] [CrossRef] [Green Version]
  41. Brink, M.; De Goeij, A.F.; Weijenberg, M.P.; Roemen, G.M.; Lentjes, M.H.; Pachen, M.M.; Smits, K.M.; De Bruïne, A.P.; Goldbohm, R.A.; van den Brandt, P. K-ras oncogene mutations in sporadic colorectal cancer in The Netherlands Cohort Study. Carcinogenesis 2003, 24, 703–710. [Google Scholar] [CrossRef] [Green Version]
  42. Ruiz-Candelaria, Y.; Miranda-Diaz, C.; Hunter-Mellado, R.F. K-RAS mutation profile in Puerto Rican patients with colorectal cancer: Trends from April 2009 to January 2011. Int. J. Biol. Markers 2013, 28, 393–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ashktorab, H.; Smoot, D.T.; Farzanmehr, H.; Fidelia-Lambert, M.; Momen, B.; Hylind, L.; Iacosozio-Dononue, C.; Carethers, J.M.; Goel, A.; Boland, C.R.; et al. Clinicopathological features and microsatellite instability (MSI) in colorectal cancers from African Americans. Int. J. Cancer 2005, 116, 914–919. [Google Scholar] [CrossRef] [Green Version]
  44. Xicola, R.M.; Gagnon, M.; Clark, J.R.; Carroll, T.; Gao, W.; Fernandez, C.; Mijic, D.; Rawson, J.B.; Janoski, A.; Pusatcioglu, C.K.; et al. Excess of Proximal Microsatellite-Stable Colorectal Cancer in African Americans from a Multiethnic Study. Clin. Cancer Res. 2014, 20, 4962–4970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Weisenberger, D.J.; Levine, A.J.; Long, T.I.; Buchanan, D.D.; Walters, R.; Clendenning, M.; Rosty, C.; Joshi, A.D.; Stern, M.C.; Le Marchand, L.; et al. Association of the Colorectal CpG Island Methylator Phenotype with Molecular Features, Risk Factors, and Family History. Cancer Epidemiology Biomarkers Prev. 2015, 24, 512–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Hughes, L.A.; Bakker, C.A.K.-D.; Smits, K.M.; Brandt, P.A.V.D.; Jonkers, D.; Ahuja, N.; Herman, J.G.; Weijenberg, M.P.; van Engeland, M. The CpG island methylator phenotype in colorectal cancer: Progress and problems. Biochim. et Biophys. Acta (BBA) –Rev. Cancer 2012, 1825, 77–85. [Google Scholar] [CrossRef]
  47. Tucker, K.; Bianchi, L.A.; Maras, J.; Bermudez, O.I. Adaptation of a Food Frequency Questionnaire to Assess Diets of Puerto Rican and Non-Hispanic Adults. Am. J. Epidemiology 1998, 148, 507–518. [Google Scholar] [CrossRef]
  48. Preston, A.M.; Palacios, C.; Rodríguez, C.A.; Vélez-Rodríguez, R.M. Validation and reproducibility of a semi-quantitative food frequency ques-tionnaire for use in the Puerto Rican population. Public Health Nutr. (submitted).
  49. Avena, S.; Via, M.; Ziv, E.; Pérez-Stable, E.J.; Gignoux, C.R.; Dejean, C.; Huntsman, S.; Torres-Mejía, G.; Dutil, J.; Matta, J.L.; et al. Heterogeneity in Genetic Admixture across Different Regions of Argentina. PLoS ONE 2012, 7, e34695. [Google Scholar] [CrossRef] [Green Version]
  50. Torres, J.B.; Kittles, R.A.; Stone, A.C. Mitochondrial and Y Chromosome Diversity in the English-Speaking Caribbean. Ann. Hum. Genet. 2007, 71, 782–790. [Google Scholar] [CrossRef]
  51. Janakiraman, M.; Vakiani, E.; Zeng, Z.; Pratilas, C.A.; Taylor, B.S.; Chitale, D.; Halilovic, E.; Wilson, M.; Huberman, K.; Filho, J.C.R.; et al. Genomic and Biological Characterization of Exon 4 KRAS Mutations in Human Cancer. Cancer Res. 2010, 70, 5901–5911. [Google Scholar] [CrossRef] [Green Version]
  52. Neumann, J.; Zeindl-Eberhart, E.; Kirchner, T.; Jung, A. Frequency and type of KRAS mutations in routine diagnostic analysis of metastatic colorectal cancer. Pathol. Res. Pr. 2009, 205, 858–862. [Google Scholar] [CrossRef] [PubMed]
  53. Hobbs, G.A.; Wittinghofer, A.; Der, C.J. Selective Targeting of the KRAS G12C Mutant: Kicking KRAS When It’s Down. Cancer Cell 2016, 29, 251–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Van Cutsem, E.; Köhne, C.-H.; Láng, I.; Folprecht, G.; Nowacki, M.P.; Cascinu, S.; Shchepotin, I.; Maurel, J.; Cunningham, D.; Tejpar, S.; et al. Cetuximab Plus Irinotecan, Fluorouracil, and Leucovorin As First-Line Treatment for Metastatic Colorectal Cancer: Updated Analysis of Overall Survival According to Tumor KRAS and BRAF Mutation Status. J. Clin. Oncol. 2011, 29, 2011–2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. De Roock, W.; Jonker, D.J.; Di Nicolantonio, F.; Sartore-Bianchi, A.; Tu, D.; Siena, S.; Lamba, S.E.; Arena, S.; Frattini, M.; Piessevaux, H.; et al. Association of KRAS p.G13D Mutation with Outcome in Patients With Chemotherapy-Refractory Metastatic Colorectal Cancer Treated With Cetuximab. JAMA 2010, 304, 1812–1820. [Google Scholar] [CrossRef] [Green Version]
  56. Tejpar, S.; Celik, I.; Schlichting, M.; Sartorius, U.; Bokemeyer, C.; Van Cutsem, E. Association of KRAS G13D Tumor Mutations With Outcome in Patients With Metastatic Colorectal Cancer Treated with First-Line Chemotherapy With or Without Cetuximab. J. Clin. Oncol. 2012, 30, 3570–3577. [Google Scholar] [CrossRef]
  57. Jones, R.P.; Sutton, P.A.; Evans, J.P.; Clifford, R.; McAvoy, A.; Lewis, J.; Rousseau, A.; Mountford, R.; McWhirter, D.; Malik, H.Z. Specific mutations in KRAS codon 12 are associated with worse overall survival in patients with advanced and recurrent colorectal cancer. Br. J. Cancer 2017, 116, 923–929. [Google Scholar] [CrossRef] [Green Version]
  58. Pérez-Mayoral, J.; Soto-Salgado, M.; Shah, E.; Kittles, R.; Stern, M.C.; Olivera, M.I.; Gonzalez-Pons, M.; Rodriguez-Quilichinni, S.; Torres, M.; Reyes, J.S.; et al. Association of genetic ancestry with colorectal tumor location in Puerto Rican Latinos. Hum. Genom. 2019, 13, 1–11. [Google Scholar] [CrossRef] [Green Version]
  59. Petrick, J.L.; Barber, L.E.; Andersen, S.W.; Florio, A.A.; Palmer, J.R.; Rosenberg, L. Racial Disparities and Sex Differences in Early- and Late-Onset Colorectal Cancer Incidence, 2001–2018. Front. Oncol. 2021, 11, 734998. [Google Scholar] [CrossRef]
  60. Thompson, A.E.; Anisimowicz, Y.; Miedema, B.; Hogg, W.; Wodchis, W.P.; Aubrey-Bassler, K. The influence of gender and other patient characteristics on health care-seeking behaviour: A QUALICOPC study. BMC Fam. Pract. 2016, 17, 12. [Google Scholar] [CrossRef] [Green Version]
  61. Gausman, V.; Dornblaser, D.; Anand, S.; Hayes, R.B.; O’Connell, K.; Du, M.; Liang, P.S. Risk Factors Associated With Early-Onset Colorectal Cancer. Clin. Gastroenterol. Hepatol. 2019, 18, 2752–2759. [Google Scholar] [CrossRef]
  62. Archambault, A.N.; Lin, Y.; Jeon, J.; Harrison, T.A.; Bishop, D.T.; Brenner, H.; Casey, G.; Chan, A.T.; Chang-Claude, J.; Figueiredo, J.C.; et al. Nongenetic Determinants of Risk for Early-Onset Colorectal Cancer. JNCI Cancer Spectr. 2021, 5, pkab029. [Google Scholar] [CrossRef] [PubMed]
Table 1. Sociodemographic and clinicopathological characteristics of the study population.
Table 1. Sociodemographic and clinicopathological characteristics of the study population.
Characteristicsn%
Age at CRC diagnosis (n = 717)
  <50 years24534.2
  ≥50 years47265.8
Gender (n = 718)
  Male37151.7
  Female34748.3
Ancestry (n = 413)
  African admixture0.21 ± 0.01
  European admixture0.61 ± 0.01
  Amerindian admixture0.18 ± 0.003
Educational Level (n = 455)
  <High school diploma or equivalent17939.3
  ≥High school diploma or equivalent27660.7
Marital Status (n = 377)
  Single/Divorced/Widowed11029.2
  Living Together/Married26770.8
Health Insurance (n = 482)
  Private17335.9
  Public15732.6
  Medicare/Medicare Advantage15231.5
Family History of CRC (n = 650)
  Yes18027.7
  No47072.3
Current Drinker (n = 663)
  No49775.0
  Yes16625.0
Current Smoker (n = 663)
  No62394.0
  Yes406.0
BMI (n = 704)
  <25 (Underweight/Normal)22231.5
  ≥25 (Overweight/Obese)48268.5
Location of CRC tumor (n = 533)
  Proximal11922.3
  Distal38772.6
  Colon, unspecified275.1
Tumor Stage (n = 708)
  I/II50971.9
  III/IV19928.1
Tumor Differentiation (n = 421)
  High11527.3
  Low/Moderate30672.7
Counts vary between the variables due to missing information.
Table 2. Frequency of tumor biomarkers among PRH according to age at CRC diagnosis.
Table 2. Frequency of tumor biomarkers among PRH according to age at CRC diagnosis.
Markers<50 Years
n (%)
≥50 Years
n (%)
p-Value
BRAF (n = 134)
Wild type39 (90.7)82 (67.8)0.999
Mutation4 (9.3)9 (9.9)
KRAS (n = 144)
Wild type28 (58.3)70 (72.9)0.077
Mutation20 (41.7)26 (27.1)
Microsatellite status (n = 192)
MSS67 (93.1)117 (97.5)0.312
MSI-Low1 (1.4)1 (0.8)
MSI-High4 (5.6)2 (1.7)
CIMP status (n = 111)
None2 (7.7)8 (9.4)0.999
Low24 (92.3)76 (89.4)
High0 (0.0)1 (1.2)
Counts vary between variables due to missing information.
Table 3. Frequency of KRAS mutations among tumors from PRH.
Table 3. Frequency of KRAS mutations among tumors from PRH.
COSMIC IDCodon AffectedHGVS cDNAHGVS Protein No. of Times Observed
COSM51612c.34G > Tp.G12C2
COSM51712c.34G > Ap.G12S0
COSM52012c.35G > Tp.G12V4
COSM52112c.35G > Ap.G12D12
COSM52212c.35G > Cp.G12A4
COSM52713c.37G > Tp.G13C3
COSM53113c.38-39GC > ATp.G13D5
COSM53213c.38G > Ap.G13D9
COSM55361c.182A > Tp.Q61L2
COSM55461c.183A > Cp.Q61H3
COSM55561c.183A > Tp.Q61H1
Table 4. Sociodemographic and clinicopathological characteristics according to CRC pathway in PRH.
Table 4. Sociodemographic and clinicopathological characteristics according to CRC pathway in PRH.
CharacteristicsPathways (n = 186)
Traditional
n (%)
Serrated
n (%)
Alternate
n (%)
Other Pathway
n (%)
p-Value
n = 38n = 14n = 42n = 92
Age at diagnosis
(n = 186)
  <50 years16 (42.1)5 (35.7)17 (40.5)38 (41.3)0.986
  ≥50 years22 (57.9)9 (64.3)25 (59.5)54 (58.7)
Gender
(n = 186)
  Male24 (63.2)4 (28.6)21 (50.0)41 (44.6)0.108
  Female14 (36.8)10 (71.4)21 (50.0)51 (55.4)
Ancestry
(n = 123)
  African Admixture0.20 ± 0.090.19 ± 0.050.24 ± 0.160.17 ± 0.070.307
  European Admixture0.62 ± 0.100.66 ± 0.050.61 ± 0.160.63 ± 0.100.582
  Amerindian Admixture0.18 ± 0.070.14 ± 0.060.15 ± 0.060.19 ± 0.070.029
Educational Level
(n = 144)
  <High school diploma or equivalent6 (19.4)4 (36.4)19 (54.3)18 (26.9)0.013
  ≥High school diploma or equivalent25 (80.7)7 (63.6)16 (45.7)49 (73.1)
Marital Status
(n = 107)
  Single/Divorced/Widowed6 (30.0)6 (54.6)11 (32.4)16 (38.1)0.549
  Living Together/Married14 (70.0)5 (45.5)23 (67.7)26 (61.9)
Health Insurance
(n = 145)
  Private19 (57.8)2 (22.2)13 (34.2)22 (33.9)0.054
  Public5 (15.2)5 (55.6)9 (23.7)25 (38.5)
  Medicare/Medicare Advantage9 (27.3)2 (22.2)16 (42.1)18 (27.7)
Family History of CRC (n = 179)
  Yes28 (75.7)12 (92.3)31 (77.5)60 (67.4)0.239
  No9 (24.3)1 (4.7)9 (22.5)29 (32.6)
Current Drinker
(n = 171)
  No25 (67.6)12 (92.1)29 (72.5)54 (66.7)0.294
  Yes12 (32.4)1 (7.7)11 (27.5)27 (33.3)
Smoker
(n = 171)
  No35 (92.1)11 (84.6)37 (94.9)75 (92.6)0.654
  Yes3 (7.9)2 (15.4)2 (5.1)6 (7.4)
BMI
(n = 185)
  <25 (Underweight/Normal)9 (23.7)3 (23.1)19 (45.2)27 (29.4)0.160
  ≥25 (Overweight/Obese)29 (76.3)10 (76.9)23 (54.8)65 (70.7)
Tumor location
(n = 152)
  Proximal10 (27.8)7 (50.0)11 (29.0)17 (26.6)0.247
  Distal25 (69.4)7 (50.0)23 (60.5)46 (71.9)
  Colon, unspecified1 (2.8)0 (0.0)4 (10.5)1 (1.6)
Tumor Stage
(n = 184)
  I/II33 (86.8)13 (92.9)36 (85.7)76 (84.4)0.940
  III/IV5 (13.2)1 (7.1)6 (14.3)14 (15.6)
Tumor Differentiation (n = 135)
  High11 (31.4)2 (16.7)10 (30.3)11 (20.0)0.511
  Low/Moderate24 (68.6)10 (83.3)23 (69.7)44 (80.0)
Counts vary between variables due to missing information.
Table 5. Sociodemographic and clinicopathological characteristics evaluated according to age at CRC diagnosis.
Table 5. Sociodemographic and clinicopathological characteristics evaluated according to age at CRC diagnosis.
Characteristic<50 Years≥50 Yearsp-Value
Gender (n = 717)
  Male102 (41.6)268 (56.8)<0.001
  Female143 (58.4)204 (43.2)
Ancestry (n = 412)
  African Admixture 0.22 ± 0.100.20 ± 0.130.056
  European Admixture0.59 ± 0.120.62 ± 0.110.009
  Amerindian Admixture0.19 ± 0.070.17 ± 0.080.009
Educational Level (n = 455)
  <High school diploma or equivalent42 (25.8)137 (46.9)<0.001
  ≥High school diploma or equivalent121 (74.2)155 (53.1)
Marital Status (n = 377)
  Single/Divorced/Widowed37 (27.6)73 (30.0)0.679
  Living Together/Married97 (72.4)170 (70.0)
Health Insurance (n = 482)
  Private89 (53.0)84 (26.8)<0.001
  Public72 (42.9)85 (27.1)
  Medicare/Medicare Advantage7 (4.2)145 (46.2)
Family History of CRC (n = 650)
  Yes76 (33.3)104 (24.6)0.018
  No152 (66.7)318 (75.4)
Current Drinker (n = 662)
  No169 (74.1)328 (75.6)0.681
  Yes59 (25.9)106 (24.4)
Current Smoker (n = 662)
  No214 (94.3)408 (93.8)0.806
  Yes13 (5.7)27 (6.21)
BMI (n = 703)
  <25 (Underweight/Normal)76 (31.3)146 (31.7)0.900
  ≥25 (Overweight/Obese)167 (68.7)314 (68.3)
Tumor location (n = 532)
  Proximal40 (21.9)79 (22.6)0.835
  Distal135 (73.8)251 (71.9)
  Colon, unspecified8 (4.4)19 (5.4)
Tumor Stage (n = 707)
  I/II169 (70.1)339 (72.8)0.462
  III/IV72 (29.9)127 (27.3)
Tumor Differentiation (n = 420)
  High39 (27.5)75 (27.0)0.916
  Low/Moderate103 (72.5)203 (73.0)
Counts vary between variables due to missing information.
Table 6. Comparison of gender, BRAF and KRAS mutation status, microsatellite instability and CIMP status among different populations.
Table 6. Comparison of gender, BRAF and KRAS mutation status, microsatellite instability and CIMP status among different populations.
Study Population
CharacteristicPRHSpanishp-ValueChinesep-ValueUSp-Value
Gender
  Male51.7% (717)59.0% (n = 876)0.00457.0% (n = 84)0.35845.0% (n = 1253)0.004
  Female48.3 % (n = 717)41.0% (n = 876)43.0% (n = 84)55.0% (n = 1253)
Markers
BRAF mutated9.7% (n = 134) 6.2% (n = 878)0.1300.7% (n = 401)<0.00114.5% (n = 1253)0.128
KRAS mutated31.9% (n = 144)36.9% (n = 878)0.24724.9% (n = 401)0.10435.9% (1249)0.342
MSI Status
  High3.1% (n = 192)9.1% (n = 878)-6.1% (n = 82)0.24615.4% (n = 1253)<0.001
  Low1.0% (n = 192) -23.2% (n = 82)<0.001
  MSS98.9% (n = 192)--70.7% (n = 82)<0.00184.6% (n = 1253)<0.001
CIMP Status
  None9.0% (n = 111)--12.5% (n = 401)0.31143.6% (n = 1173)<0.001
  Low90.0% (n = 111)--77.3% (n = 401)0.00338.9% (n = 1173)<0.001
  High0.9% (n = 111)27.4% (n = 878)<0.00110.2% (n = 401)0.00217.5% (n = 1173)<0.001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Perez-Mayoral, J.; Gonzalez-Pons, M.; Centeno-Girona, H.; Montes-Rodríguez, I.M.; Soto-Salgado, M.; Suárez, B.; Rodríguez, N.; Colón, G.; Sevilla, J.; Jorge, D.; et al. Molecular and Sociodemographic Colorectal Cancer Disparities in Latinos Living in Puerto Rico. Genes 2023, 14, 894. https://doi.org/10.3390/genes14040894

AMA Style

Perez-Mayoral J, Gonzalez-Pons M, Centeno-Girona H, Montes-Rodríguez IM, Soto-Salgado M, Suárez B, Rodríguez N, Colón G, Sevilla J, Jorge D, et al. Molecular and Sociodemographic Colorectal Cancer Disparities in Latinos Living in Puerto Rico. Genes. 2023; 14(4):894. https://doi.org/10.3390/genes14040894

Chicago/Turabian Style

Perez-Mayoral, Julyann, Maria Gonzalez-Pons, Hilmaris Centeno-Girona, Ingrid M. Montes-Rodríguez, Marievelisse Soto-Salgado, Belisa Suárez, Natalia Rodríguez, Giancarlo Colón, Javier Sevilla, Daphne Jorge, and et al. 2023. "Molecular and Sociodemographic Colorectal Cancer Disparities in Latinos Living in Puerto Rico" Genes 14, no. 4: 894. https://doi.org/10.3390/genes14040894

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop