Skip to main content
AACR Open Access logoLink to AACR Open Access
. 2025 Jun 27;34(9):1472–1482. doi: 10.1158/1055-9965.EPI-24-1711

The Translational Research Program in Cancer Differences across Populations

Jane C Figueiredo 1,#,*, Diana Redwood 2,#, Li Li 3, Elizabeth Donato 4, Daniel Fort 3, Eric E Fox 5, William M Grady 4, Heather Green 3, Tabitha A Harrison 6, Carl Haupt 3, Li Hsu 4, Meredith AJ Hullar 4, Jeroen R Huyghe 4, Wenora Johnson 5, Amanda L Koehne 4, Scott D LaBrie 4, Meredith A Lakey 3, MingGang Lin 4, Nicole C Loroña 1, Grace A Maresh 3, Marc Matrana 3, Jonathan D Mizrahi 3, Sarah H Nash 7, Nathalie T Nguyen 1, Jennifer L Paruch 3, Amanda I Phipps 6, Conghui Qu 4, Timothy W Randolph 4, Stephanie Romo 1, Claire E Thomas 4, Sushma Thomas 4, James Tiesinga 2, Charles Whitlow 3, Cecilia CS Yeung 4, Hang Yin 4, Craig M Zibilich 3, Christopher I Li 4,, Timothy K Thomas 2,, Ulrike Peters 4,‡,*
PMCID: PMC12349377  NIHMSID: NIHMS2094161  PMID: 40576634

Abstract

Background:

Cancer incidence and mortality vary substantially across populations. The Translational Research Program in Cancer Differences across Populations (TRPCDP) was established in 2020 to address differences in cancer incidence and mortality rates within the United States, with a particular focus on colorectal cancer.

Methods:

The TRPCDP centralized data acquisition and harmonization across three sites in the United States to create a well-annotated resource of colorectal cancer tumors across four populations: African American/Black, Alaska Native, Hispanic/Latino/Latina, and non-Hispanic White. Using a case–control framework, patients with lethal colorectal cancer were matched to two controls with nonlethal colorectal cancer. Formalin-fixed, paraffin-embedded tumor and normal tissue were retrieved and sent for centralized pathology review, followed by DNA and RNA extraction and tissue microarray development. Multiomics and spatial profiling are underway to evaluate the transcriptome, proteome, and microbiome. Patient demographic and clinical data were obtained by medical record review, patient self-report, or linkage to cancer registries. Additional health-related factors were assessed using geospatial linkage.

Results:

The virtual biorepository includes 7,181 patients [African American (n = 1,345), Alaska Native (n = 1,640), Hispanic (n = 1,659), and non-Hispanic White (n = 2,537)]. Tissue blocks (1,594 tumor and 728 normal colon samples) were selected for 938 patients. To date, DNA and RNA have been extracted (n = 831), and tissue microarrays have been constructed (n = 414). Transcriptomic analysis, spatial tumor profiling (multiplex immunofluorescence, PhenoCycler, and GeoMx), and microbiome data (16S rRNA sequencing and digital droplet PCR) are available.

Conclusions:

The TRPCDP has developed a clinically annotated biorepository for future molecular epidemiology studies.

Impact:

The TRPCDP is a unique program that supports collaborative research, community engagement, and pipeline development for the next generation of scientists.

Introduction

Colorectal cancer is a preventable disease, yet it remains a common cancer worldwide. In 2024, colorectal cancer became the leading cause of cancer mortality in men and the second leading cause in women <50 years of age (1) For multifactorial reasons that remain unresolved, colorectal cancer disproportionately affects some communities (2). This is especially true among Alaska Native peoples, who have incidence rates that are more than double US national rates (3). These high rates are a concern for Alaska Native communities, who are working to reduce the burden through increased colorectal cancer screening and prevention. African American/Black individuals (herein referred to as African American) have the second highest incidence and mortality rates for colorectal cancer in the United States (20% and 40% higher than the general US population; ref. 4). Although rates are lower than average among adults in the Hispanic/Latino/Latina (herein referred to as Hispanic) community, this population has experienced the lowest decline in mortality rates over recent decades compared with any other population (5). These communities also experience a high proportion of early-onset colorectal cancer, which has been increasing in incidence since the mid-1990s (2).

Variation in incidence and mortality rates across populations underscores the importance of investigating the complex interplay of environmental and biological factors contributing to cancer-related differences in risk and outcomes. However, few resources exist with sufficient representation from multiple communities (6, 7). Inadequate sample sizes have limited studies published to date but show results that merit further investigation, including differences in the expression of clinically actionable tumor markers when stratified by demographic factors including geography, race, and ethnicity (810), reduced treatment efficacy in some genetically defined populations (11, 12), and altered gut microbiota across populations (13) with implications for treatment efficacy (14). These observed differences may reflect underlying genetic diversity, as well as varying environmental exposures and other factors (15, 16). Ongoing innovations in biotechnology and the emergence of novel bioinformatic and machine learning algorithms offer tremendous hope for developing better predictive biomarkers of treatment response and understanding the complex biological contributors that explain differences in the cancer burden.

The Translational Research Program in Cancer Differences across Populations (TRPCDP) was established to provide the necessary infrastructure for applying novel biotechnologies and analytic methods to identify molecular and biological drivers of clinical utility and inform the development of new therapeutic strategies across different populations. Herein, we describe the rationale and development of TRPCDP. By studying population groups with varying cancer rates, we enhance our ability to identify the molecular characteristics and contributing factors driving these differences. Our overarching goal is to meaningfully reduce morbidity and mortality across populations, by (i) discovery and validation of novel molecular, biological, and microbial changes related to response to treatment and risk of lethal cancer and (ii) development of clinical interventions aimed at reducing cancer risk and mortality.  While the current focus of TRPCDP is in on colorectal cancer our long term goal is to expand the work to other cancer types with varying incidence and mortality rates across populations. We describe the organizational structure and methodology of TRPCDP, including the biorepository inventory, current and pending projects, community stakeholder engagement and participation, training of the next generation of cancer researchers, and the creation of an accessible resource for future research.

Materials and Methods

Overview

The foundational work of TRPCDP began with collaborative efforts established more than a decade ago, including forming a consortium with data pooled from the Colon Cancer Family Registry (CCFR; ref. 17), Colorectal Cancer Transdisciplinary Study (CORECT; refs. 18, 19), Genetics and Epidemiology of Colorectal Cancer Consortium (GECCO; refs. 18, 20, 21), ColoCare (22), and the Hispanic Colorectal Cancer Study (HCCS; refs. 23, 24). In 2020, the Fred Hutchinson Cancer Center (FHCC; Seattle, WA), in collaboration with the Alaska Native Tribal Health Consortium (ANTHC; Anchorage, AK), Cedars-Sinai Medical Center (Los Angeles, CA), and Ochsner Health (New Orleans, LA) initiated the TRPCDP (Fig. 1). The TRPCDP is supported by an NCI P20 feasibility and planning grant (P20CA252733) and NCI P50 Specialized Programs of Research Excellence (P50CA285275) and through subsequent support through foundations (V Foundation and Goldman Sachs Foundation), FHCC, Cedars-Sinai, Ochsner Health, ANTHC, and investigator-initiated NCI grants (R01CA155101, R01CA238087, R01CA284732, R01CA248931, R01CA280639, and U01CA290673).

Figure 1.

Figure 1.

Organizational structure of the Translational Research Program in Cancer Differences across Populations based on the P20 “mini-SPORE”. Depiction of the population groups included in the TRPCDP and the program organizational structure: executive committee; advisory boards; biospecimen, data and pathology core; research projects; administrative core; and developmental research and career enhancement programs. (Created with BioRender.com.)

The four populations currently represented in TRPCDP are Alaska Native, Hispanic, non-Hispanic White, and African American recruited from the following regions in the United States. The study is conducted in accordance with the Declaration of Helsinki, Council for International Organizations of Medical Sciences (CIOMS) guidelines, the Belmont Report, and the US Common Rule.

Alaska

ANTHC is a nonprofit tribal health organization, owned, managed, and operated by Alaska Native peoples, which provides cancer care and treatment primarily at the Alaska Native Medical Center (ANMC) located in Anchorage, AK, to the approximately 151,000 Alaska Native people living in Alaska. Since 2000, more than 1,600 Alaska Native people living in Alaska have been diagnosed with colorectal cancer (3). American Indian people who have moved to Alaska are able to get care at the ANMC and are included in Surveillance, Epidemiology, and End Results Program (SEER) data. Formalin-fixed, paraffin-embedded (FFPE) tumor blocks going back to the 1970s at the ANMC Pathology Lab Archive were available for TRPCDP use.

This portion of the study was reviewed and approved by the Alaska Area Institutional Review Board (IRB; IRB00000636), which operates under Federalwide Assurance FWA00001138.

California

The HCCS was established in 2010 as a population-based cohort of Hispanic individuals using the California Cancer Registry and direct access to local hospitals. More than 1,600 Hispanic patients with colorectal cancer have been enrolled in the HCCS (2426). FFPE blocks were retrieved directly from hospitals throughout California. Patient-reported surveys, medical chart abstraction, and data from linkages to the SEER California Cancer Registry and the National Death Index are available. The Cedars-Sinai Medical Center’s IRB (FWA00000468) reviewed and approved this study, and all participants signed informed consent.

Louisiana

Ochsner Health is the largest healthcare provider in Louisiana, serving more than 1.4 million patients per year. It has a long tradition of serving a large African American population, and about one third of its patients identify as African American. Data from medical chart abstraction and linkages with the Louisiana SEER registry/National Death Index for demographic, clinical, outcome, and treatment data are available. Since 2000, Ochsner has diagnosed and treated more than 1,300 African American and more than 2,500 non-Hispanic White patients with colorectal cancer. FFPE blocks were retrieved from their centralized tissue bank and the Department of Pathology. The Ochsner IRB approved this project.

Selection of TRPCDP participants

Patients diagnosed with colorectal cancer at ANMC or Ochsner Health, as well as research participants in the HCCS, were eligible for inclusion. Additional samples were also included from participants in the Seattle Colon Cancer Family Registry (see the Collaborations section). Eligible participants were diagnosed with colon or rectal adenocarcinoma (ICD10 codes: C180 and C182–C189 for colon adenocarcinoma; C199 and C209 for rectal adenocarcinoma; ref. 27) after 2000 and of 18 to 85 years of age at the time of diagnosis. An upper limit of 85 years of age was selected because the outcome of interest was colorectal cancer–specific mortality. Individuals >85 years of age have many competing causes of death and may not receive standard treatment. Focusing on stage I to III also ensured greater homogeneity of first-line therapy. From this, we constructed a virtual biorepository of all participants with colorectal cancer with information on age at diagnosis, sex, tumor stage, and site (Supplementary Table S1).

TRPCDP study design and sample size targets

To optimize statistical power, TRPCDP utilized a nested case–control design in which cases were defined as patients who died of colorectal cancer, and controls were patients with colorectal cancer who survived at least as long as the case to which they were matched. Controls were matched to cases 2:1 on race and ethnicity, age at diagnosis, sex, stage, tumor site, and year of diagnosis. The goal was ∼70 cases of lethal colorectal cancer and ∼140 matched controls for each population group.

Biospecimen collection and processing

Centralized pathology review and tissue handling

All tissue samples (FFPE blocks and/or unstained slides) were retrieved by staff at the participating sites and subsequently shipped to the FHCC. All tissues were reviewed centrally by a single pathologist (ALK) using a standard operating procedure with additional pathologists available for second opinions as needed. Each FFPE tissue block was evaluated for quality of tissue preservation and processing, histology, confirmation of correct diagnosis, tumor content, and tumor volume using an hematoxylin and eosin (H&E) slide to ensure sufficient material was available for DNA and RNA extraction and annotation of tumor tissue selection for tissue microarray (TMA) generation (Fig. 2).

Figure 2.

Figure 2.

Sample sharing and analysis workflow for TRPCDP, 2020–2023. Evaluation of FFPE tumor and normal tissue blocks for quality of tissue preservation and processing, histology, confirmation of correct diagnosis, and tumor content and volume prior to TMA construction. MSI, microsatellite instability. (Created with BioRender.com.)

DNA and RNA extraction

To ensure at least 70% tumor content for DNA and RNA extraction from FFPE tissue sections and unstained slides, tumor areas were marked on the corresponding H&E-stained slide to guide dissection of tumor tissue. All paraffin was removed by treatment with the Deparaffinization Solution (Qiagen). RNA extraction was done using the Qiagen RNeasy FFPE kit (Qiagen) according to the manufacturer’s protocol. Extraction of microbial DNA followed a standard protocol that was adapted for FFPE tissue elsewhere (28, 29) Tumor DNA and RNA and microbial DNA are maintained centrally at the FHCC.

Transcriptomic and microbiome analyses

RNA sequencing (RNA-seq) libraries were generated using the Illumina TruSeq RNA Exome kit (Illumina, Inc.). Libraries were sequenced on NovaSeq 6000 to a minimum depth of 30 million paired reads per sample. Microbial DNA was analyzed using 16S rRNA gene sequencing to classify the relative abundance of different bacterial genera and phyla as well as α and β diversity in both normal and tumor tissue. We developed and applied digital droplet PCR (ddPCR) assays to quantify the following three microbes with putative links to colorectal cancer: Fusobacterium nucleatum, enterotoxigenic Bacteroides fragilis, polyketide synthase positive Escherichia coli and the human SLCO2A1 gene in FFPE human tissue.

TMA construction

To ensure data robustness, we used three tumor cores per tissue, two collected from the center of the tumor and one from the invasive margin of the tumor. In addition, we collected one core from the normal epithelial tissue. Core selection from FFPE blocks and unstained slides was guided by TRPCDP pathologist (ALK) review of H&E-stained slides. TMAs were constructed using the TMA Grand Master (3DHISTECH). Tissue cores from case–control pairs were distributed throughout a TMA to minimize batch effects. TMAs are maintained centrally at the FHCC.

Multiomics profiling

We profiled a subset of TMAs on various platforms: multiplex immunofluorescence, Akoya PhenoCycler, RNAscope, 10× Visium spatial gene expression, NanoString GeoMx digital spatial profiling and NanoString CosMx.

Covariates

We centralized variable definition and a minimal set of variables for abstraction to ensure uniform collection at each participating site. Across all sites, data were collected using the secure Health Insurance Portability and Accountability Act–compliant Research Electronic Data Capture (REDCap) online data entry system adapted from the ColoCare study (22). The FHCC team, which also oversees the abstraction in ColoCare, provided training to TRPCDP abstractors to ensure a high-quality, standardized approach across sites.

Medical record abstraction

Trained medical abstractors used standardized abstraction forms adapted from the ColoCare Study (22) to manually review medical records for detailed demographic (sex, age, race, and ethnicity), clinical (body mass index, comorbidities, surgical procedures, and treatments), and epidemiologic (smoking and alcohol) data. Patient records were evaluated five years prior to and after colorectal cancer diagnosis including clinic notes, pathology reports, radiology reports, operative/procedure reports, hospital discharge summaries, radiation and medical oncology notes, laboratory results, tumor recurrence, and outpatient summaries (Table 1).

Table 1.

Data elements collected and harmonized in Translational Research Program in Cancer Differences across Populations.

Domain Variable Source
Demographic Age, date of birth, sex, race, and ethnicity Cancer registries, medical records, patient surveys, and linkage to external databases (geocoding)
Medical history Family history of cancer, comorbid conditions, prescription medication use, height, weight, smoking history, and pre-CRC surgery antibiotics Medical records and patient surveys
Tumor characteristics Grade, histology, clinical/pathologic stage, and lymphovascular and perineural invasion Cancer registries and medical records
Molecular testing Microsatellite status, BRAF and KRAS mutations, and other testing Medical records and molecular profiling
Surgical treatment Type and dates Cancer registries and medical records
Radiotherapy Dates, number of treatments, and dose Cancer registries and medical records
Chemotherapy Agents, dose, dates, and number of infusions Cancer registries and medical records
Immunotherapy Agents, dose, dates, and number of infusions Cancer registries and medical records
Treatment side effects Allergic reactions, fatigue, cardiac/GI/hematologic/neurologic toxicities, and other adverse events Medical records
Clinical outcomes CRC recurrences (dates, location, and methods of detection and treatment), progression-free survival, vital status, and cause of death (1°, 2°, and 3° causes) Cancer registries and medical records
Access to and use of healthcare Insurance status, insurance type, screening history, and primary care utilization Cancer registries and medical records

NOTE: Data from cancer registry and linkage to external databases (geocoding) are available for all patients in the virtual repository (Supplementary Table S1), survey data for all Hispanic patients (Supplementary Table S1), and medical record collection is ongoing for all patients in the physical repository (Table 2).

Abbreviations: CRC, colorectal cancer; GI, gastrointestinal.

Linkage to cancer registries

We obtained data from SEER cancer registries on the following clinical variables: age at diagnosis, sex, tumor site, stage, surgery, chemotherapy, radiation, recurrence, overall colorectal cancer–specific death, and age of death. We compared data obtained from medical records and adjudicated discrepancies.

Patient-reported surveys

HCCS participants completed demographic, lifestyle, and medical history questionnaires including colorectal cancer–specific outcomes like recurrence. Data were used to verify and augment information from medical records and cancer registries as needed.

Other health-related factors

Where available, we leveraged data from SEER cancer registries and geospatial linkage that assessed factors such as rurality or geographic access to care based on the patients’ geocoded addresses at the time of diagnosis (3036).

Collaborations

The TRPCDP is actively engaging with international collaborators to pool additional data on participants with colorectal cancer.

Seattle Colon Cancer Family Registry

The Seattle Colon Cancer Family Registry (SCCFR) is a population-based case–control study in Washington State, one of seven centers forming the Colon Cancer Family Registry (www.coloncfr.org). Recruitment of cases and controls began in 1998 and ended in 2012. The SCCFR collected information on demographics, risk factors, family, medical, and reproductive histories, physical activity, diet, survival, and tumor characteristics. Through an ongoing ancillary study within the SCCFR, data were also collected to characterize the tumor-associated microbiome in colorectal cancer, using 16S rRNA-seq and ddPCR assays for F. nucleatum on paired tumor and adjacent normal colon tissue samples. From within this data resource, a sample of 146 non-Hispanic White participants meeting eligibility for TRPCDP analyses was selected and pooled with other included populations for analysis.

Administration

TRPCDP administration functions were centralized at the FHCC to provide programmatic support; coordinate activities within the biospecimen and pathology core to provide sample processing and data management services, coordinate laboratory analyses, and establish future collaborations; build a development research program to support pilot studies; and coordinate activities of the community, internal, and external advisory boards (Fig. 1). We established standard protocols, material transfer agreements, and data transfer agreements between the TRPCDP partners and the FHCC to facilitate the secure transfer of samples and related data.

Community Advisory Board

Given the focus on colorectal cancer differences in incidence and mortality by race and ethnicity and our primary goal of conducting translational research that meaningfully reduces cancer rates, our Community Advisory Board consisted of African American, Alaska Native, and Hispanic patient advocates and key community stakeholders. These individuals shared their lived experience with colorectal cancer, patient advocacy work, and provided critical feedback to inform research priorities, research design, implementation, and dissemination of study results.

Scientific internal and external advisory boards

Internal and External Advisory Board members were chosen to provide experience in leading large program grants and scientific expertise in the areas of colorectal cancer research, clinical medicine, cancer disparity research, and community-based participatory research. Board members represented a range of backgrounds, including current NCI P20 Cancer Disparities principal investigators, NCI P50 Specialized Programs of Research Excellence principal investigators, FHCC institutional leadership, and researchers from TRPCDP-participating institutions.

Data availability

We developed a framework for sharing data in accordance with the FAIR principles: Findable, Accessible, Interoperable, and Reusable (37). Biospecimens and data collected as part of TRPCDP were available to all TRPCDP pilot project awardees. After prioritizing TRPCDP projects and pilots, biospecimen and data will be made available through non-TRPCDP investigators after appropriate approvals are in place. Data will also be made publicly available via controlled access, such as the NIH Database of Genotypes and Phenotypes. As TRPCDP involves samples from Alaska Native peoples who have tribal sovereignty over their data and biospecimens, we developed a rigorous review and approval process for requesting data (including via the NIH Database of Genotypes and Phenotypes data repository) in collaboration with the NCI, the Alaska Area IRB, and Alaska tribal research review committees. Data from Alaska Native peoples are subject to tribal sovereignty and have additional review and approval requirements for their use (Supplemental Fig. S1).

Results

Characteristics of TRPCDP study participants

Currently, data and biospecimens from a total of 938 (African American: 214; Alaska Native: 319; Hispanic: 185; and non-Hispanic White: 220) patients with colorectal cancer are available for molecular assessments in the TRPCDP physical repository (Table 2; Supplementary Table S2). Study characteristics are reflective of the selection criteria (see Selection of TRPCDP participants) and, as such, vary from those in the virtual repository. The mean age at diagnosis (64.1 years ± 11.9) was similar across population groups, except for the Hispanic cohort, which had a lower mean age at diagnosis (57.8 ± 12.3). A nearly equal representation of females to males is available across all population groups (48%). Approximately one fifth of patients had rectal cancers (17%), with a higher proportion in the Hispanic population (31%). There were more patients with colorectal cancer tumors at more advanced stage of diagnosis [stage I: 190 (20%); stage II: 351 (37%); stage III: 397 (42%)]; however, the distribution varies by ethnicity and race [African American: stage I: 36 (17%), stage II: 82 (38%), and stage III: 96 (45%); Alaska Native: stage I: 56 (18%), stage II: 126 (39%), and stage III: 137 (43%); Hispanic: stage I: 43 (23%), stage II: 56 (30%), and stage III: 86 (46%); and non-Hispanic White: stage I: 25 (25%), stage II: 40 (40%), and stage III: 35 (35%)]. Per the case–control design, one third of all patients had lethal colorectal cancer. The physical repository will continue to grow based on ongoing and future funding. For instance, current efforts will increase the number of patients with early-onset colorectal cancer and patients undergoing immune checkpoint inhibitor treatment.

Table 2.

Summary of Translational Research Program in Cancer Differences across Populations current physical repository stratified by population group.

African American Alaska Native Hispanic Non-Hispanic White
N % N % N % N %
Patients with colorectal cancer 214 319 185 220
Diagnosis years 2000–2015 2000–2017 2008–2016 2000–2015
Age at diagnosis (years)
 Mean 64.2 65.2 57.8 67.9
 <50 21 10% 34 11% 45 24% 14 6%
 50–60 52 24% 71 22% 62 34% 45 21%
 >60 141 66% 214 67% 78 42% 161 73%
Female 107 50% 195 61% 77 42% 78 35%
Tumor stage
 I 36 17% 56 18% 43 23% 55 25%
 II 82 38% 126 39% 56 30% 87 40%
 III 96 45% 137 43% 86 47% 78 35%
 IV NA
Tumor site
 Proximal colon 101 47% 161 50% 60 32% 109 50%
 Distal colon 87 41% 124 39% 63 34% 61 28%
 Rectum 24 11% 33 10% 57 31% 47 21%
 Unspecified 2 1% 1 1% 7 3% 3 1%
 Tumor tissue # of blocks 401 537 248 408
 Normal tissue # of blocks 204 245 79 200
 Extracted DNA and RNA 206 258 153 214
 TMA 113 111 69 121

TRPCDP biorepository

As of August 2024, the biorepository includes 2,322 FFPE tissue blocks (1,594 tumor and 728 normal tissues from the surgical margin). Due to funding for additional research projects, the largest number of tissue blocks is available for the Alaska Native group: 554 tumor and 253 normal blocks with DNA/RNA extracted for 217 patients with colorectal cancer and TMAs constructed for 112 patients with colorectal cancer. In the African American cohort, there are 421 tumor and 212 normal blocks available, with DNA/RNA extracted for 212 patients with colorectal cancer and TMAs constructed for 118 patients with colorectal cancer. The non-Hispanic White cohort contains 412 tumor and 201 normal blocks with DNA/RNA extraction for 218 patients with colorectal cancer and TMAs for 120 patients with colorectal cancer. The Hispanic cohort is the smallest with 248 tumor and 66 normal blocks and DNA/RNA extracted for 151 patients with colorectal cancer and TMAs constructed for 62 patients with colorectal cancer.

TRPCDP projects and pilot studies

Starting in 2020, there were multiple projects which used biospecimens from TRPCDP for risk prediction, tumor sequencing, transcriptomics, tumor microbiome analysis, spatial profiling, and treatment response studies, including comparison of population groups in terms of colorectal cancer–specific survival as well as gene expression of tumor tissues and aspects of the microbial community in tumors (Table 3). Specimens from 782 patients with colorectal cancer underwent RNA-seq, and 715 underwent microbiome assessments. A total of 10 TMAs, representing 123 patients, have undergone GeoMx and PhenoCycler analyses so far. Another funded ancillary study will develop a cohort of Alaska Native individuals undergoing colorectal cancer screening to examine risk factors for colorectal cancer in this population and build an additional biorepository for future research (U01CA290673).

Table 3.

Translational Research Program in Cancer Differences across Populations Projects, 2020–2023.

Project Technology Population Number of samples
Main P20 projects
 Identifying novel molecular predictors of lethal CRC RNA-seq AA, AN, Hispanic, and NHW 840
 Examine the CRC-associated microbiome using candidate and agnostic approaches 16S rRNA-seq and ddPCR AA, AN, Hispanic, and NHW 840
Developmental pilot projects
 Predictive factors in CRC progression and treatment response mIF AA and NHW 30
 Metabolic dependencies in obesity-associated CRCs
  • CRISPR/Cas-9, in vivo mouse studies, and mass spectrometry

Not applicable (animal study)
 Developing single-cell omics from colorectal mucosa biopsies 10X Single Cell Multiome NHW 4
 Characterizing the CRC tumor microenvironment PhenoCycler AN 13
 Testing a microbiome-modulating fiber supplement in rectal cancer
  • ddPCR, 16S rRNA-seq, and RNAscope

AA and NHW 12
 Gene-level correlates of the gut microbiome and colorectal neoplasia Shotgun sequencing AN 30
 Prognostic biomarker evaluation in patients with pancreatic cancer
  • RNA ISH and multiplex IHC

AA and NHW 40
 Measuring the CRC intratumoral mycobiome in diverse racial and ethnic populations 18S rRNA-seq AA, AN, Hispanic, and NHW 40
 A DNA microarray approach to characterize the intratumoral microbiome DNA microarray AA, AN, Hispanic, and NHW 40
Technical feasibility studies
  •  NanoString GeoMx digital spatial profiler

  •  10X Genomics Visium spatial transcriptomics

  •  PhenoCycler for senescence markers

  • GeoMx

  • Visium

  • PhenoCycler

AN

Abbreviations: AA, African American; AN, Alaska Native; CRC, colorectal cancer; mIF, multiplex immunofluorescence; NHW, non-Hispanic White.

Discussion

The overarching goal of TRPCDP is to use molecular epidemiology studies to identify biological factors that drive colorectal cancer incidence and mortality, which may explain observed population differences. By examining population groups with distinct colorectal cancer rates, we can increase the range of molecular characteristics affecting colorectal cancer development and mortality and thereby increase the statistical power. We developed an organizational infrastructure representative of four populations with stark differences in colorectal cancer incidence and mortality rates, built a rich annotated biorepository, initiated collaborative research projects, established a process for community engagement and participation in research, and created the resources and a training environment for the next generation of cancer researchers. Our major scientific priorities center around clinically relevant questions in colorectal cancer differences in outcomes. The main themes and how our program is designed to advance our major scientific priorities are discussed below:

  • 1. Inclusion of communities rarely studied in cancer research: Most of the focus in cancer research has been on investigating factors driving non-Hispanic White and African American differences (10, 3841). The TRPCDP addresses a critical gap in knowledge for both admixed populations including the Hispanic community and Alaska Native peoples. Studies of Alaska Native populations are particularly scarce, and most have focused on implementation research aimed at improving screening (4249). These studies successfully illustrated increases in screening utilization among Alaska Native peoples to a level that now mirrors rates in the general US population (4251), leading us to consider how to make similar progress in addressing the high mortality rates in this less studied population. Similarly, for the Hispanic community, our group has led efforts to investigate genetic susceptibility and the somatic and immune landscape of colorectal tumors (15, 1921, 23, 5255). TRPCDP is poised to make significant strides in dissecting the underlying biological drivers of colorectal cancer mortality across multiple populations, as this is a first-of-its-kind infrastructure that has centralized data/biospecimens across four populations.

  • 2. Identification and validation of biological drivers influencing and reflecting differences in colorectal cancer mortality across populations: The Cancer Genome Atlas revolutionized the field of cancer genomics but was constrained by a significant gap in representation across populations, which may mean missing the full spectrum of variation. To date, we and others have demonstrated that tumors from African American patients exhibit distinct mutational frequencies in several key colorectal cancer genes, including PIK3CA, KRAS, and BRAF, as well as differences in the expression of genes associated with inflammation and immune response (41). Fewer than 20% of those included in the Human Tumor Atlas Network identify as African American or Hispanic, and the number of Alaska Native and American Indian patients with cancer is too small to be listed separately. Consequently, The Cancer Genome Atlas and Human Tumor Atlas Network have limited ability to provide accurate tumor characterization for the full breadth of populations affected by colorectal cancer. The TRPCDP aims to fill this gap by investigating population-based differences in colorectal cancer incidence and mortality. Through this research, we aim to create a comprehensive profile of the colorectal cancer tumor transcriptome and microbiome across four distinct populations. Comparing data across groups with vastly different incidence and mortality rates may advance our understanding of molecular and biological drivers and markers of differences in colorectal cancer rates. We will also increase the predictive performance of molecular and microbial markers and the identification of novel therapeutic targets/approaches, through the inclusion of more individuals of varying background and exposures. Given observed differences in disease burden across these population groups, such inclusion can also inform the development of novel strategies, particularly among populations with reduced access to medical services. Published work in other cancers demonstrates the effectiveness of this approach in identifying novel, clinically meaningful prognostic and predictive tumor features (5660). By expanding this methodology to colorectal cancer, we aim to uncover critical insights that can lead to significant improvements in patient outcomes across populations.

  • 3. Spotlight on the intratumoral microbiota across populations: A growing body of evidence has implicated the gut microbiota’s role in colorectal cancer progression through multiple mechanisms that include enhancement of host pro-inflammatory genes and cytokines (6163), induction of oxidative stress (64, 65), affecting the immune system, and via the action of bacteria-derived genotoxins (6669). However, to date, studies of the tumor microbiome in colorectal cancer have been conducted mainly in non-Hispanic White populations. However, the microbiome could play an important role in observed differences in colorectal cancer rates across population groups given differences in lifestyle and environmental exposures (e.g., diet and antibiotic use). Although a growing number of studies are exploring the gut microbiome using stool samples (61, 7073), less data are available on the tumor microbiome (7476). Identifying aspects of, and patterns in, the intratumoral microbiome may provide avenues to enhance prediction of incident colorectal cancer and adverse outcomes that ultimately lead to improvements in colorectal cancer survival. In the TRPCDP, we propose using discovery-based, agnostic, and targeted candidate approaches to identify differences and similarities in the tumor-associated microbiome across populations. To agnostically compare patterns of tumor-associated microbial community structure, we sequence bacterial 16S rRNA gene segments from tumor tissue specimens and matched normal tissue specimens. Furthermore, we developed and applied highly sensitive and quantitative ddPCR assays to compare the quantities of candidate bacteria in tumors across populations. This approach aims to identify potential novel microbial biomarkers and pathways that may be associated with or contribute to differences in colorectal cancer outcomes.

  • 4. Tumor tissue–based spatially resolved predictors of treatment response across populations: We propose a highly innovative colorectal cancer tumor molecular profiling strategy. Specifically, spatial proteomic analyses enable deep characterization of the tumor microenvironment and immune response, which in other cancers has been shown to be of high translational relevance (7779). Few studies, however, have applied this powerful approach to colorectal cancer or advanced our understanding of cancer by studying patients from multiple populations. It is critical to identify new biomarkers, given that carcinoembryonic antigen (CEA), the most commonly used biomarker clinically to assess disease progression, has only moderate sensitivity and specificity for detecting recurrent colorectal cancer. Its use remains controversial as a Cochrane review of CEA’s utility concluded that “CEA is insufficiently sensitive to be used alone” and that it is “essential to augment CEA monitoring with another diagnostic modality in order to avoid missed cases (recurrences; ref. 80).” Thus, there is an urgent need for novel biomarkers that can be used to guide clinical decision-making and that perform well among populations of patients that experience the greatest colorectal cancer burden. Through this initiative, we aim to develop biomarkers that are not only more sensitive and specific than current options but that perform optimally in all individuals. This will enhance personalized treatment strategies, reduce differences in colorectal cancer outcomes, and ultimately lead to improved survival and quality of life.

  • 5. Bi-directional partnership with community stakeholders: For the successful translation of our research, community engagement is key, as it can result in higher-quality, more generalizable, and more acceptable research. Our Community Advisory Board, comprising patient advocates, is a key TRPCDP partner, providing critical feedback and actively engaging in the execution of our research and the dissemination of study results. Community engagement improves the effectiveness of our communication with diverse audiences (81), improving our ability to disseminate information across various health literacy levels and cultural preferences (82).

  • 6. Preparing a new generation of cancer researchers: TRPCDP plans to mentor and support early career investigators and established faculty members interested in careers on translational cancer research through research support, leadership development, and enhanced recruitment and retention strategies at TRPCDP-participating institutions.

Our study has strengths and limitations. Most importantly, our program represents multiple groups across the United States that experience high as well as low colorectal cancer incidence rates. None of the studies harmonized in the TRPCDP include individuals who self-identify as Asian or other combinations of race and ethnicity beyond the four described above; however, there exists several notable resources including the International Cancer Genome Consortium and Accelerating Research in Genomic Oncology, 100,000 Genomes Project (83, 84), and others (8588) that will allow opportunities for collaboration in the future. When faced with the need to develop a well-powered study with sufficient follow-up time in a timely and cost-effective manner, reliance on archival specimens and data is crucial. Archival FFPE tissue blocks have become increasingly valuable for research, given that a growing number of molecular profiling platforms can utilize FFPE tissue, allowing for detailed tumor profiling at the single-cell level. All patient recruitment areas are covered by SEER cancer registries, which have enabled us to start with a well-harmonized virtual repository of key patient characteristics. This allowed us to select the most informative patients and design a well-balanced and matched set of patients for downstream analysis. Enriching our analysis for patients who died of colorectal cancer substantially increases the power for our primary outcome of colorectal cancer–specific death. However, not all patients selected from the virtual repository have usable tumor tissue; for approximately 20% of patients, the tissue blocks are either unavailable or have too low tumor content for extraction and analysis. To account for this and the case–control sampling approach, we use inverse probability weighting to their sampling fractions derived from available SEER data, which provides complete information on the underlying patient population. Another limitation is the lack of comprehensive risk factor data, including dietary variables, in the TRPCDP, given limitations of electronic medical records. Linkage to other databases and leveraging geospatial data will help increase data on various health-related factors (89).

In summary, high-dimensional molecular and microbiome data in large, well-characterized patients derived from different population groups are urgently needed. These data are essential to improve statistical power and to develop better prediction models to inform prognosis. It could also help guide the selection of therapeutic agents and the development of novel strategies. Through this integrated effort, we envision realizing our goal of a sustained translational research program dedicated to eliminating differences in colorectal cancer incidence and mortality rates across populations and, more broadly, reducing colorectal cancer–related morbidity and mortality in the Unites States.

Supplementary Material

Supplemental Figure 1

Supplemental Figure 1. Shows the process for requesting data from Alaska Native peoples from the Alaska Native Tribal Health Consortium and accessible via the NIH dbGaP data repository.

Supplemental Table 1

Supplemental Table 1 shows a Summary of Translational Research Program in Colorectal Cancer Disparities virtual repository of colorectal cancer patients.

Supplemental Table 2

Supplemental Table 2 Shows a summary of Translational Research Program in Colorectal Cancer Disparities physical repository of colorectal cancer patients stratified by case-control status

Acknowledgments

We acknowledge the research participants of projects conducted by the TRPCDP, the contributions and support of the Alaska Native Tribal Health Consortium and the Southcentral Foundation Board of Directors for review and approval of this manuscript, and the staff of the TRPCDP-participating institutions for their work on the study. The findings and conclusions in this article are those of the authors and do not necessarily represent the official position of the NIH. This research was supported by several funding sources, including the NIH NCI grant P20CA252733: J.C. Figueiredo, D. Redwood, L. Li, E. Donato, W.M. Grady, H. Green, T.A. Harrison, L. Hsu, M.A.J. Hullar, J.R. Huyghe, A.L. Koehne, M. Lin, G.A. Maresh, N.T. Nguyen, A.I. Phipps, C. Qu, T.W. Randolph, S. Romo, C.E. Thomas, S. Thomas, U. Peters, C.I. Li, and T.K. Thomas; grant P50CA285275: D Redwood, L. Li, E. Donato, W.M. Grady, H. Green, T.A. Harrison, L. Hsu, M.A.J. Hullar, J.R. Huyghe, A.L. Koehne, M. Lin, G.A. Maresh, M. Matrana, J.D. Mizrahi, N.T. Nguyen, A.I. Phipps, C. Qu, T.W. Randolph, S. Romo, C.E. Thomas, S. Thomas, C.C.S. Yeung, U. Peters, C.I. Li, T.K. Thomas, and J.C. Figueiredo; grant R01CA280639: U. Peters, D. Redwood, E. Donato, L. Hsu, A.L. Koehne, M. Lin, C. Qu, S. Thomas, and J.R. Huyghe; grants R01CA155101, R01CA238087, R01CA284732, and R01CA248931: J.C. Figueiredo; grant U01CA290673: D. Redwood, U. Peters, and C.I. Li; Goldman Sachs Foundation: U. Peters, M.A.J. Hullar, L. Hsu, M. Lin, G.A. Maresh, C. Qu, T.K. Thomas, L. Li, D. Redwood, S. Thomas, C.E. Thomas, J.R. Huyghe, and C.I. Li; and V Foundation: U. Peters, L. Hsu, M. Lin, C. Qu, J.R. Huyghe, T.K. Thomas, S. Thomas, D. Redwood, C.E. Thomas, and C.I. Li. This research also benefited from the support of shared resources: Experimental Histopathology Shared Resource (RRID: SCR_022612); Genomics and Bioinformatics Shared Resource (RRID: SCR_022606); and Biospecimen Processing and Biorepository Shared Resource: part of the Fred Hutchinson Cancer Center/University of Washington/Seattle Children’s Cancer Consortium (P30 CA015704). Furthermore, the V Foundation contributed to the support of this research.

Footnotes

Note: Supplementary data for this article are available at Cancer Epidemiology, Biomarkers & Prevention Online (http://cebp.aacrjournals.org/).

Authors’ Disclosures

J.C. Figueiredo reports grants from NIH during the conduct of the study. D. Redwood reports grants from NIH during the conduct of the study. W.M. Grady reports personal fees from Guardant Health and Karius outside the submitted work. M. Matrana reports personal fees from Merck, Eisai, AstraZeneca, Johnson and Johnson/Janssen, Exelixis, EMD Serono, Pfizer, Astellas Pharma, Bristol Myers Squibb, Seagen, and Genentech outside the submitted work. J.D. Mizrahi reports personal fees from Seagen, Daiichi Sankyo, AstraZeneca, Exelixis, Amgen, Bristol Myers Squibb, and Intera Oncology outside the submitted work. S. Thomas reports grants from NIH during the conduct of the study. C.C.S. Yeung reports personal fees from TwinStrand, Thermo Fisher Scientific, and Beckman Coulter and grants from Sensei Bio, OBI Pharma, and Kineta outside the submitted work. U. Peters reports other support from AbbVie during the conduct of the study; reports being a consultant for AbbVie; and reports that her husband is holding individual stocks for the following companies: Amazon, ARM Holdings plc, BioNTech, BYD Company Limited, CrowdStrike Holdings, Inc., CureVac, Google/Alphabet, Microsoft Corporation, NVIDIA Corporation, and Stellantis. No disclosures were reported by the other authors.

Authors’ Contributions

J.C. Figueiredo: Conceptualization, resources, investigation, methodology, writing–original draft, writing–review and editing. D. Redwood: Conceptualization, resources, investigation, methodology, writing–original draft, project administration, writing–review and editing. L. Li: Resources, methodology, writing–review and editing. E. Donato: Writing–review and editing. D. Fort: Methodology, writing–original draft, writing–review and editing. E.E. Fox: Methodology, writing–review and editing. W.M. Grady: Methodology, writing–review and editing. H. Green: Methodology, writing–review and editing. T.A. Harrison: Writing–review and editing. C. Haupt: Writing–review and editing. L. Hsu: Methodology, writing–review and editing. M.A.J. Hullar: Methodology, writing–review and editing. J.R. Huyghe: Methodology, writing–review and editing. W. Johnson: Writing–review and editing. A.L. Koehne: Methodology, writing–review and editing. S.D. LaBrie: Writing–review and editing. M.A. Lakey: Resources, project administration, writing–review and editing. M. Lin: Resources, project administration, writing–review and editing. N.C. Loroña: Writing–review and editing. G.A. Maresh: Resources, writing–review and editing. M. Matrana: Writing–review and editing. J.D. Mizrahi: Resources, writing–review and editing. S.H. Nash: Writing–review and editing. N.T. Nguyen: Resources, project administration, writing–review and editing. J.L. Paruch: Writing–review and editing. A.I. Phipps: Resources, writing–review and editing. C. Qu: Writing–review and editing. T.W. Randolph: Writing–original draft, writing–review and editing. S. Romo: Resources, project administration, writing–review and editing. C.E. Thomas: Writing–review and editing. S. Thomas: Resources, project administration, writing–review and editing. J. Tiesinga: Writing–review and editing. C. Whitlow: Resources, writing–review and editing. C.C.S. Yeung: Resources, writing–review and editing. H. Yin: Writing–review and editing. C.M. Zibilich: Writing–original draft, writing–review and editing. C.I. Li: Conceptualization, resources, funding acquisition, methodology, project administration, writing–review and editing. T.K. Thomas: Conceptualization, resources, funding acquisition, methodology, project administration, writing–review and editing. U. Peters: Resources, supervision, funding acquisition, investigation, writing–original draft, project administration, writing–review and editing.

References

  • 1. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin 2024;74:12–49. [DOI] [PubMed] [Google Scholar]
  • 2. Rahman R, Schmaltz C, Jackson CS, Simoes EJ, Jackson-Thompson J, Ibdah JA. Increased risk for colorectal cancer under age 50 in racial and ethnic minorities living in the United States. Cancer Med 2015;4:1863–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Zimpelman GL, Miller KN, Carlo DD, Schade TL, Provost EM, Britton CL, et al. Cancer in Alaska native people: 1969–2018, the 50-year report. Anchorage, AK: Alaska Native Tumor Registry, Alaska Native Epidemiology Center, Alaska Native Tribal Health Consortium 2021;27:1–185. [Google Scholar]
  • 4. American Cancer Society . Cancer facts & figures for African Americans 2019-2021. [Cited 2019 Aug 2024]. Available from:https://www.cancer.org/research/cancer-facts-statistics/cancer-facts-figures-for-african-americans.html.
  • 5. American Cancer Society . Colorectal cancer facts & figures 2023-2025. 2023[Cited 2024 Mar 11]. Available from:https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/colorectal-cancer-facts-and-figures/colorectal-cancer-facts-and-figures-2023.pdf.
  • 6. Knepper TC, McLeod HL. When will clinical trials finally reflect diversity? Nature 2018;557:157–9. [DOI] [PubMed] [Google Scholar]
  • 7. Jones N, Marks R, Ramirez R, Rios-Vargas M. 2020 census illuminates racial and ethnic composition of the country. Washington (DC): United States Census Bureau; 2021[Cited 2023 Jan 9]. Available from:https://www.census.gov/library/stories/2021/08/improved-race-ethnicity-measures-reveal-united-states-population-much-more-multiracial.html. [Google Scholar]
  • 8. Yoon HH, Shi Q, Alberts SR, Goldberg RM, Thibodeau SN, Sargent DJ, et al. Racial differences in BRAF/KRAS mutation rates and survival in stage III colon cancer patients. J Natl Cancer Inst 2015;107:djv186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Staudacher JJ, Yazici C, Bul V, Zeidan J, Khalid A, Xia Y, et al. Increased frequency of KRAS mutations in African Americans compared with caucasians in sporadic colorectal cancer. Clin Transl Gastroenterol 2017;8:e124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Heath EI, Lynce F, Xiu J, Ellerbrock A, Reddy SK, Obeid E, et al. Racial disparities in the molecular landscape of cancer. Anticancer Res 2018;38:2235–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Lee SHR, Antillon-Klussmann F, Pei D, Yang W, Roberts KG, Li Z, et al. Association of genetic ancestry with the molecular subtypes and prognosis of childhood acute lymphoblastic leukemia. JAMA Oncol 2022;8:354–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Yang JJ, Cheng C, Devidas M, Cao X, Fan Y, Campana D, et al. Ancestry and pharmacogenomics of relapse in acute lymphoblastic leukemia. Nat Genet 2011;43:237–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Dwiyanto J, Hussain MH, Reidpath D, Ong KS, Qasim A, Lee SWH, et al. Ethnicity influences the gut microbiota of individuals sharing a geographical location: a cross-sectional study from a middle-income country. Sci Rep 2021;11:2618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Björk JR, Bolte LA, Maltez Thomas A, Lee KA, Rossi N, Wind TT, et al. Longitudinal gut microbiome changes in immune checkpoint blockade-treated advanced melanoma. Nat Med 2024;30:785–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Zavala VA, Bracci PM, Carethers JM, Carvajal-Carmona L, Coggins NB, Cruz-Correa MR, et al. Cancer health disparities in racial/ethnic minorities in the United States. Br J Cancer 2021;124:315–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. American Assciation for Cancer Research . AACR cancer disparities report 2024; 2024[Cited 2025 Feb 20]. Available from:https://cancerprogressreport.aacr.org/wp-content/uploads/sites/2/2024/05/AACR_CDPR__2024.pdf.
  • 17. Newcomb PA, Baron J, Cotterchio M, Gallinger S, Grove J, Haile R, et al. Colon Cancer Family Registry: an international resource for studies of the genetic epidemiology of colon cancer. Cancer Epidemiol Biomarkers Prev 2007;16:2331–43. [DOI] [PubMed] [Google Scholar]
  • 18. Fernandez-Rozadilla C, Timofeeva M, Chen Z, Law P, Thomas M, Schmit S, et al. Deciphering colorectal cancer genetics through multi-omic analysis of 100,204 cases and 154,587 controls of European and east Asian ancestries. Nat Genet 2023;55:89–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Schmit SL, Edlund CK, Schumacher FR, Gong J, Harrison TA, Huyghe JR, et al. Novel common genetic susceptibility loci for colorectal cancer. J Natl Cancer Inst 2019;111:146–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Huyghe JR, Bien SA, Harrison TA, Kang HM, Chen S, Schmit SL, et al. Discovery of common and rare genetic risk variants for colorectal cancer. Nat Genet 2019;51:76–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Thomas M, Su Y-R, Rosenthal EA, Sakoda LC, Schmit SL, Timofeeva MN, et al. Combining Asian and European genome-wide association studies of colorectal cancer improves risk prediction across racial and ethnic populations. Nat Commun 2023;14:6147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Ulrich CM, Gigic B, Böhm J, Ose J, Viskochil R, Schneider M, et al. The colocare study: a paradigm of transdisciplinary science in colorectal cancer outcomes. Cancer Epidemiol Biomarkers Prev 2019;28:591–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Ricker CN, Hanna DL, Peng C, Nguyen NT, Stern MC, Schmit SL, et al. DNA mismatch repair deficiency and hereditary syndromes in Latino patients with colorectal cancer. Cancer 2017;123:3732–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Schmit SL, Schumacher FR, Edlund CK, Conti DV, Ihenacho U, Wan P, et al. Genome-wide association study of colorectal cancer in Hispanics. Carcinogenesis 2016;37:547–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Black DS, Li MJ, Ihenacho U, Nguyen NT, de Fatima Reyes M, Milam J, et al. Shared health characteristics in Hispanic colorectal cancer patients and their primary social support person following primary diagnosis. Psychooncology 2016;25:1028–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Black DS, Lam CN, Nguyen NT, Ihenacho U, Figueiredo JC. Complementary and integrative health practices among hispanics diagnosed with colorectal cancer: utilization and communication with physicians. J Altern Complement Med 2016;22:473–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. World Health Organization . ICD-10: international statistical classification of diseases and related health problems. 10th Rev. 2nd ed. [Cited 2004 Sep 6]. Available from:https://apps.who.int/iris/handle/10665/42980.
  • 28. Hullar MAJ, Jenkins IC, Randolph TW, Curtis KR, Monroe KR, Ernst T, et al. Associations of the gut microbiome with hepatic adiposity in the multiethnic cohort adiposity Phenotype study. Gut Microbes 2021;13:1965463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 2017;358:1443–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Yu M, Tatalovich Z, Gibson JT, Cronin KA. Using a composite index of socioeconomic status to investigate health disparities while protecting the confidentiality of cancer registry data. Cancer Causes Control 2014;25:81–92. [DOI] [PubMed] [Google Scholar]
  • 31. Moss JL, Stinchcomb DG, Yu M. Providing higher resolution indicators of rurality in the surveillance, epidemiology, and end results (SEER) database: implications for patient privacy and research. Cancer Epidemiol Biomarkers Prev 2019;28:1409–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Yost K, Perkins C, Cohen R, Morris C, Wright W. Socioeconomic status and breast cancer incidence in California for different race/ethnic groups. Cancer Causes Control 2001;12:703–11. [DOI] [PubMed] [Google Scholar]
  • 33. Ellis L, Canchola AJ, Spiegel D, Ladabaum U, Haile R, Gomez SL. Racial and ethnic disparities in cancer survival: the contribution of tumor, sociodemographic, institutional, and neighborhood characteristics. J Clin Oncol 2018;36:25–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Kish JK, Yu M, Percy-Laurry A, Altekruse SF. Racial and ethnic disparities in cancer survival by neighborhood socioeconomic status in Surveillance, Epidemiology, and End Results (SEER) Registries. J Natl Cancer Inst Monogr 2014;2014:236–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Henry KA, Sherman RL, McDonald K, Johnson CJ, Lin G, Stroup AM, et al. Associations of census-tract poverty with subsite-specific colorectal cancer incidence rates and stage of disease at diagnosis in the United States. J Cancer Epidemiol 2014;2014:823484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Lewis-Thames MW, Langston ME, Khan S, Han Y, Fuzzell L, Xu S, et al. Racial and ethnic differences in rural-urban trends in 5-year survival of patients with lung, prostate, breast, and colorectal cancers: 1975-2011 surveillance, epidemiology, and end results (SEER). JAMA Netw Open 2022;5:e2212246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Wilkinson MD, Dumontier M, Aalbersberg IJJ, Appleton G, Axton M, Baak A, et al. The FAIR Guiding Principles for scientific data management and stewardship. Sci Data 2016;3:160018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Al Abo M, Hyslop T, Qin X, Owzar K, George DJ, Patierno SR, et al. Differential alternative RNA splicing and transcription events between tumors from African American and White patients in The Cancer Genome Atlas. Genomics 2021;113:1234–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Wang Z, Li L, Guda K, Chen Z, Barnholtz-Sloan J, Park YS, et al. Adverse clinical outcome associated with mutations that typify African American colorectal cancers. J Natl Cancer Inst 2016;108:djw164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Ashktorab H, Azimi H, Varma S, Lee EL, Laiyemo AO, Nickerson ML, et al. Driver genes exome sequencing reveals distinct variants in African Americans with colorectal neoplasia. Oncotarget 2019;10:2607–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Jovov B, Araujo-Perez F, Sigel CS, Stratford JK, McCoy AN, Yeh JJ, et al. Differential gene expression between African American and European American colorectal cancer patients. PLoS One 2012;7:e30168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Nash SH, Britton C, Redwood D. Characteristics of colorectal cancers among Alaska Native people before and after implementing programs to promote screening. J Cancer Pol 2021;29:100293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Seeff LC, Major A, Townsend JS, Provost E, Redwood D, Espey D, et al. Comprehensive cancer control programs and coalitions: partnering to launch successful colorectal cancer screening initiatives. Cancer Causes Control 2010;21:2023–31. [DOI] [PubMed] [Google Scholar]
  • 44. Redwood D, Provost E, Perdue D, Haverkamp D, Espey D. The last frontier: innovative efforts to reduce colorectal cancer disparities among the remote Alaska Native population. Gastrointest Endosc 2012;75:474–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Redwood D, Joseph DA, Christensen C, Provost E, Peterson VL, Espey D, et al. Development of a flexible sigmoidoscopy training program for rural nurse practitioners and physician assistants to increase colorectal cancer screening among Alaska Native people. J Health Care Poor Underserved 2009;20:1041–8. [DOI] [PubMed] [Google Scholar]
  • 46. Muller CJ, Robinson RF, Smith JJ, Jernigan MA, Hiratsuka V, Dillard DA, et al. Text message reminders increased colorectal cancer screening in a randomized trial with Alaska Native and American Indian people. Cancer 2017;123:1382–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Redwood DG, Blake ID, Provost EM, Kisiel JB, Sacco FD, Ahlquist DA. Alaska native patient and provider perspectives on the multitarget stool DNA test compared with colonoscopy for colorectal cancer screening. J Prim Care Community Health 2019;10:2150132719884295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Redwood DG, Dinh TA, Kisiel JB, Borah BJ, Moriarty JP, Provost EM, et al. Cost-effectiveness of multitarget stool DNA testing vs colonoscopy or fecal immunochemical testing for colorectal cancer screening in Alaska native people. Mayo Clin Proc 2021;96:1203–17. [DOI] [PubMed] [Google Scholar]
  • 49. Alaska Department of Health and Social Services . Behavioral risk factor surveillance system (BRFSS). [Cited 2020 Sep 6]. Available from:https://health.alaska.gov/dph/Chronic/Pages/brfss/default.aspx.
  • 50. Joseph DA, King JB, Richards TB, Thomas CC, Richardson LC. Use of colorectal cancer screening tests by state. Prev Chronic Dis 2018;15:E80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. American Cancer Society . Cancer facts & figures 2022. Atlanta: American Cancer Society. [Cited 2022 Aug 12]. Available from:https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2022.html. [Google Scholar]
  • 52. Matejcic M, Teer JK, Hoehn HJ, Diaz DB, Shankar K, Gong J, et al. Colorectal tumors in diverse patient populations feature a spectrum of somatic mutational profiles. Cancer Res 2025;85:1928–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Matejcic M, Teer JK, Hoehn HJ, et al. Genetic ancestry and somatic mutational frequencies in colorectal cancer: findings from the Latino Colorectal Cancer Consortium. Cancer Res 2020;4:1–31. [Google Scholar]
  • 54. Schmit SL, Purrington K, Figueiredo JC. Efforts to grow genomic research in ancestrally diverse and admixed populations. Cancer Res 2023;83:2443–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Stern MC, Fejerman L, Das R, Setiawan VW, Cruz-Correa MR, Perez-Stable EJ, et al. Variability in cancer risk and outcomes within US latinos by national origin and genetic ancestry. Curr Epidemiol Rep 2016;3:181–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Pagès F, Mlecnik B, Marliot F, Bindea G, Ou F-S, Bifulco C, et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 2018;391:2128–39. [DOI] [PubMed] [Google Scholar]
  • 57. Schürch CM, Bhate SS, Barlow GL, Phillips DJ, Noti L, Zlobec I, et al. Coordinated cellular neighborhoods orchestrate antitumoral immunity at the colorectal cancer invasive front. Cell 2020;182:1341–59.e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Pelka K, Hofree M, Chen JH, Sarkizova S, Pirl JD, Jorgji V, et al. Spatially organized multicellular immune hubs in human colorectal cancer. Cell 2021;184:4734–52.e20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Väyrynen JP, Haruki K, Lau MC, Väyrynen SA, Ugai T, Akimoto N, et al. Spatial organization and prognostic significance of NK and NKT-like cells via multimarker analysis of the colorectal cancer microenvironment. Cancer Immunol Res 2022;10:215–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Phillips D, Matusiak M, Gutierrez BR, Bhate SS, Barlow GL, Jiang S, et al. Immune cell topography predicts response to PD-1 blockade in cutaneous T cell lymphoma. Nat Commun 2021;12:6726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 2013;14:207–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, Langella P, et al. Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS One 2011;6:e16393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Mima K, Nishihara R, Qian ZR, Cao Y, Sukawa Y, Nowak JA, et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut 2016;65:1973–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Goodwin AC, Destefano Shields CE, Wu S, Huso DL, Wu X, Murray-Stewart TR, et al. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc Natl Acad Sci U S A 2011;108:15354–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Huycke MM, Moore D, Joyce W, Wise P, Shepard L, Kotake Y, et al. Extracellular superoxide production by Enterococcus faecalis requires demethylmenaquinone and is attenuated by functional terminal quinol oxidases. Mol Microbiol 2001;42:729–40. [DOI] [PubMed] [Google Scholar]
  • 66. Gopalakrishnan V, Helmink BA, Spencer CN, Reuben A, Wargo JA. The influence of the gut microbiome on cancer, immunity, and cancer immunotherapy. Cancer Cell 2018;33:570–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe 2013;14:195–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Huycke MM, Abrams V, Moore DR. Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA. Carcinogenesis 2002;23:529–36. [DOI] [PubMed] [Google Scholar]
  • 69. Moncrief JS, Obiso R, Barroso LA, Kling JJ, Wright RL, Van Tassell RL, et al. The enterotoxin of Bacteroides fragilis is a metalloprotease. Infect Immun 1995;63:175–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Ahn J, Sinha R, Pei Z, Dominianni C, Wu J, Shi J, et al. Human gut microbiome and risk for colorectal cancer. J Natl Cancer Inst 2013;105:1907–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Toprak NU, Yagci A, Gulluoglu BM, Akin ML, Demirkalem P, Celenk T, et al. A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer. Clin Microbiol Infect 2006;12:782–6. [DOI] [PubMed] [Google Scholar]
  • 72. Farhana L, Antaki F, Murshed F, Mahmud H, Judd SL, Nangia-Makker P, et al. Gut microbiome profiling and colorectal cancer in African Americans and Caucasian Americans. World J Gastrointest Pathophysiol 2018;9:47–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Amitay EL, Werner S, Vital M, Pieper DH, Höfler D, Gierse I-J, et al. Fusobacterium and colorectal cancer: causal factor or passenger? Results from a large colorectal cancer screening study. Carcinogenesis 2017;38:781–8. [DOI] [PubMed] [Google Scholar]
  • 74. Flanagan L, Schmid J, Ebert M, Soucek P, Kunicka T, Liska V, et al. Fusobacterium nucleatum associates with stages of colorectal neoplasia development, colorectal cancer and disease outcome. Eur J Clin Microbiol Infect Dis 2014;33:1381–90. [DOI] [PubMed] [Google Scholar]
  • 75. Viljoen KS, Dakshinamurthy A, Goldberg P, Blackburn JM. Quantitative profiling of colorectal cancer-associated bacteria reveals associations between fusobacterium spp., enterotoxigenic Bacteroides fragilis (ETBF) and clinicopathological features of colorectal cancer. PLoS One 2015;10:e0119462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Allali I, Delgado S, Marron PI, Astudillo A, Yeh JJ, Ghazal H, et al. Gut microbiome compositional and functional differences between tumor and non-tumor adjacent tissues from cohorts from the US and Spain. Gut Microbes 2015;6:161–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Danenberg E, Bardwell H, Zanotelli VRT, Provenzano E, Chin S-F, Rueda OM, et al. Breast tumor microenvironment structures are associated with genomic features and clinical outcome. Nat Genet 2022;54:660–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Hickey JW, Haist M, Horowitz N, Caraccio C, Tan Y, Rech AJ, et al. T cell-mediated curation and restructuring of tumor tissue coordinates an effective immune response. Cell Rep 2023;42:113494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Enfield KSS, Colliver E, Lee C, Magness A, Moore DA, Sivakumar M, et al. Spatial architecture of myeloid and T cells orchestrates immune evasion and clinical outcome in lung cancer. Cancer Discov 2024;14:1018–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Nicholson BD, Shinkins B, Pathiraja I, Roberts NW, James TJ, Mallett S, et al. Blood CEA levels for detecting recurrent colorectal cancer. Cochrane Database Syst Rev 2015;2015:CD011134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Holt CL, Hussain A, Wachbroit R, Scott J. Precision medicine across the cancer continuum: implementation and implications for cancer disparities. JCO Precis Oncol 2017;1:1–5. [DOI] [PubMed] [Google Scholar]
  • 82. Freimuth VS, Quinn SC. The contributions of health communication to eliminating health disparities. Am J Public Health 2004;94:2053–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Cornish AJ, Gruber AJ, Kinnersley B, Chubb D, Frangou A, Caravagna G, et al. The genomic landscape of 2,023 colorectal cancers. Nature 2024;633:127–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. ICGC ARGO data platform. 2024[cited 2024 Aug 13]. Available from:https://platform.icgc-argo.org/.
  • 85. Dashti H, Dehzangi I, Bayati M, Breen J, Beheshti A, Lovell N, et al. Integrative analysis of mutated genes and mutational processes reveals novel mutational biomarkers in colorectal cancer. BMC Bioinformatics 2022;23:138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Zheng R, Zhang K, Tan S, Gao F, Zhang Y, Xu W, et al. Exosomal circLPAR1 functions in colorectal cancer diagnosis and tumorigenesis through suppressing BRD4 via METTL3-eIF3h interaction. Mol Cancer 2022;21:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Du M, Gu D, Xin J, Peters U, Song M, Cai G, et al. Integrated multi-omics approach to distinct molecular characterization and classification of early-onset colorectal cancer. Cell Rep Med 2023;4:100974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Chiang S-F, Huang H-H, Tsai W-S, Chin-Ming Tan B, Yang C-Y, Huang P-J, et al. Comprehensive functional genomic analyses link APC somatic mutation and mRNA-miRNA networks to the clinical outcome of stage-III colorectal cancer patients. Biomed J 2022;45:347–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Boehmer U, Potter J, Clark MA, Ozonoff A, Ceballos RM, Winter M, et al. Neighborhood characteristics and colorectal cancer survivors’ quality of care. Health Equity 2019;3:619–27. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Figure 1

Supplemental Figure 1. Shows the process for requesting data from Alaska Native peoples from the Alaska Native Tribal Health Consortium and accessible via the NIH dbGaP data repository.

Supplemental Table 1

Supplemental Table 1 shows a Summary of Translational Research Program in Colorectal Cancer Disparities virtual repository of colorectal cancer patients.

Supplemental Table 2

Supplemental Table 2 Shows a summary of Translational Research Program in Colorectal Cancer Disparities physical repository of colorectal cancer patients stratified by case-control status

Data Availability Statement

We developed a framework for sharing data in accordance with the FAIR principles: Findable, Accessible, Interoperable, and Reusable (37). Biospecimens and data collected as part of TRPCDP were available to all TRPCDP pilot project awardees. After prioritizing TRPCDP projects and pilots, biospecimen and data will be made available through non-TRPCDP investigators after appropriate approvals are in place. Data will also be made publicly available via controlled access, such as the NIH Database of Genotypes and Phenotypes. As TRPCDP involves samples from Alaska Native peoples who have tribal sovereignty over their data and biospecimens, we developed a rigorous review and approval process for requesting data (including via the NIH Database of Genotypes and Phenotypes data repository) in collaboration with the NCI, the Alaska Area IRB, and Alaska tribal research review committees. Data from Alaska Native peoples are subject to tribal sovereignty and have additional review and approval requirements for their use (Supplemental Fig. S1).


Articles from Cancer Epidemiology, Biomarkers & Prevention are provided here courtesy of American Association for Cancer Research

RESOURCES