Next Article in Journal
Evaluation of Autonomic Nervous System Dysfunction in Childhood Obesity and Prader–Willi Syndrome
Previous Article in Journal
Residual Humidity in Paraffin-Embedded Tissue Reduces Nucleic Acid Stability
 
 
ijms-logo
Article Menu

Article Menu

Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Brief Report

Do GWAS-Identified Risk Variants for Chronic Lymphocytic Leukemia Influence Overall Patient Survival and Disease Progression?

by
Antonio José Cabrera-Serrano
1,2,
José Manuel Sánchez-Maldonado
1,2,
Rob ter Horst
3,
Angelica Macauda
4,
Paloma García-Martín
5,
Yolanda Benavente
6,7,8,
Stefano Landi
9,
Alyssa Clay-Gilmour
10,
Yasmeen Niazi
11,12,
Blanca Espinet
13,14,
Juan José Rodríguez-Sevilla
15,
Eva María Pérez
5,
Rossana Maffei
16,
Gonzalo Blanco
13,14,
Matteo Giaccherini
9,
James R. Cerhan
17,
Roberto Marasca
16,
Miguel Ángel López-Nevot
18,
Tzu Chen-Liang
19,
Hauke Thomsen
20,
Irene Gámez
19,
Daniele Campa
9,
Víctor Moreno
6,21,22,
Silvia de Sanjosé
6,7,
Rafael Marcos-Gragera
7,23,24,25,
María García-Álvarez
26,
Trinidad Dierssen-Sotos
7,27,
Andrés Jerez
28,
Aleksandra Butrym
29,
Aaron D. Norman
17,
Mario Luppi
16,
Susan L. Slager
30,31,
Kari Hemminki
32,33,
Yang Li
34,35,
Sonja I. Berndt
36,
Delphine Casabonne
6,7,
Miguel Alcoceba
26,
Anna Puiggros
13,14,
Mihai G. Netea
34,37,
Asta Försti
11,12,
Federico Canzian
4 and
Juan Sainz
1,2,7,38,*
add Show full author list remove Hide full author list
1
Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain
2
Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain
3
CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
4
Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
5
Hospital Campus de la Salud, PTS, 18016 Granada, Spain
6
Catalan Institute of Oncology, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain
7
Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), University of Barcelona, 08908 Barcelona, Spain
8
CIBER Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
9
Department of Biology, University of Pisa, 56126 Pisa, Italy
10
Department of Epidemiology & Biostatistics, Arnold School of Public Health, University of South Carolina, Greenville, SC 29208, USA
11
Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
12
Hopp Children’s Cancer Center (KiTZ), 69120 Heidelberg, Germany
13
Molecular Cytogenetics Laboratory, Pathology Department, Hospital del Mar, 08003 Barcelona, Spain
14
Translational Research on Hematological Neoplasms Group, Cancer Research Program, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain
15
Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
16
Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
17
Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
18
Immunology Department, Virgen de las Nieves University Hospital, 18014 Granada, Spain
19
Hematology Department, Morales Meseguer University Hospital, 30008 Murcia, Spain
20
MSB Medical School Berlin, D-14197 Berlin, Germany
21
Cancer Prevention and Control Program, Unit of Biomarkers and Susceptibility, Bellvitge Biomedical Research Institute (IDIBELL), Catalan Institute of Oncology, 08907 Barcelona, Spain
22
Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, 08907 Barcelona, Spain
23
Epidemiology Unit and Girona Cancer Registry, Oncology Coordination Plan, Department of Health, Autonomous Government of Catalonia, Catalan Institute of Oncology, Girona Biomedical Research Institute (IdiBGi), 17190 Girona, Spain
24
Department of Nursing, Universitat de Girona, 17007 Girona, Spain
25
Josep Carreras Leukemia Research Institute, 08916 Girona, Spain
26
Department of Hematology, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
27
Faculty of Medicine, University of Cantabria, 39011 Santander, Spain
28
Department of Hematology, Experimental Hematology Unit, Vall d’Hebron Institute of Oncology (VHIO), University Hospital Vall d’Hebron, 08035 Barcelona, Spain
29
Department of Cancer Prevention and Therapy, Medical University of Wrocław, 50-556 Wrocław, Poland
30
Division of Computational Genomics, Mayo Clinic, Rochester, MN 85054, USA
31
Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA
32
Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
33
Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, 30605 Pilsen, Czech Republic
34
Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
35
Centre for Individualised Infection Medicine (CiiM) & TWINCORE, Joint Ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
36
Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
37
Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
38
Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada (UGR), 18012 Granada, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(9), 8005; https://doi.org/10.3390/ijms24098005
Submission received: 12 March 2023 / Revised: 14 April 2023 / Accepted: 23 April 2023 / Published: 28 April 2023

Abstract

:
Chronic lymphocytic leukemia (CLL) is the most common leukemia among adults worldwide. Although genome-wide association studies (GWAS) have uncovered the germline genetic component underlying CLL susceptibility, the potential use of GWAS-identified risk variants to predict disease progression and patient survival remains unexplored. Here, we evaluated whether 41 GWAS-identified risk variants for CLL could influence overall survival (OS) and disease progression, defined as time to first treatment (TTFT) in a cohort of 1039 CLL cases ascertained through the CRuCIAL consortium. Although this is the largest study assessing the effect of GWAS-identified susceptibility variants for CLL on OS, we only found a weak association of ten single nucleotide polymorphisms (SNPs) with OS (p < 0.05) that did not remain significant after correction for multiple testing. In line with these results, polygenic risk scores (PRSs) built with these SNPs in the CRuCIAL cohort showed a modest association with OS and a low capacity to predict patient survival, with an area under the receiver operating characteristic curve (AUROC) of 0.57. Similarly, seven SNPs were associated with TTFT (p < 0.05); however, these did not reach the multiple testing significance threshold, and the meta-analysis with previous published data did not confirm any of the associations. As expected, PRSs built with these SNPs showed reduced accuracy in prediction of disease progression (AUROC = 0.62). These results suggest that susceptibility variants for CLL do not impact overall survival and disease progression in CLL patients.

1. Introduction

Chronic lymphocytic leukemia (CLL) is the most common form of leukemia among adults worldwide [1], and its global health burden has risen substantially over the past 30 years [2]. CLL remains an incurable disease [3] with a heterogeneous clinical course and a 10-year survival rate ranging from 47.3% to 72.5% in males and 58.2% to 78.7% in females [4]. Traditional clinical prognostic factors include Rai and Binet staging systems, lymphocyte doubling time, cytogenetic alterations, and point mutations, which are used for patient risk stratification and clinical management [5]. Although age, sex, exposure to chemicals, race/ethnicity, and family history of hematological cancers influence the risk of CLL, recent studies have suggested that the combination of these classical factors with genetic markers might help in predicting disease onset and clinical outcome [6]. However, despite the overall success of genome-wide association studies (GWAS) in identifying susceptibility loci for CLL [7,8,9,10], there remains an unmet need to characterize genetic markers associated with disease progression and overall patient survival. Considering this background, we investigated whether GWAS-identified susceptibility variants for CLL could influence the overall survival (OS) of CLL patients and their disease progression, defined as time to first treatment (TTFT). Finally, we explored whether the effect of selected variants on OS and TTFT could be mediated by immune-related processes through a comprehensive battery of functional experiments developed in the 500FG cohort recruited in the context of the Human Functional Project (HFGP).

2. Results

CLL patients from the CRuCIAL cohort had a mean age of 65.87 and a male/female ratio of 1.57, which is in line with the worldwide median age of diagnosis and gender distributions (Table 1) [11]. Median follow-up time was 76.77 months without variation of follow-up statistics for censored patients, and the total number of deceased patients was 287. The OS did not differ significantly by country, ruling out the possibility of any deviation due to multicenter randomized patient recruitment. On the other hand, CLL patients with TTFT data in the CRuCIAL cohort had a mean age of 65.03 and a male/female ratio of 1.62. The median time to first treatment was 759.49 days and the number of deceased patients was 111 (Table 1).
Cox regression analyses showed that ten genetic variants within the CAMK2D, CASP8, CFLAR, CXXC1, GPR37, IRF8, LEF1, MYNN, PRKD2, and TERC loci were associated with OS at p < 0.05 level (Table 2). Although potentially interesting, none of these associations remained significant after correction for multiple testing, which suggested a weak effect (if any) of these genes in determining patient survival. The lack of previous studies assessing the impact of GWAS-identified risk variants on OS hampered the performance of eventual meta-analyses.
As expected, we found a weak association between the weighted and unweighted PRSs and OS of CLL patients (HR = 1.22, p = 1.80 × 10−5 and HRScaled_80% = 1.19, p = 7.61 × 10−5). Weighted and unweighted PRSs increased the capacity to predict OS by only 6–7%, with an area under the receiver operating characteristic curve (AUROC) for the unweighted and weighted PRS of 0.56 and 0.57, respectively (Table 3).
In agreement with these findings, we found that none of these SNPs were correlated with host immune parameters (cQTL data, absolute numbers of 91 blood-derived cell populations, 106 serum immunological proteins, or 7 steroid hormones), which reinforced the hypothesis of a null effect of these markers in determining overall patient survival.
On the other hand, Cox regression analyses adjusted for age, sex, and country of origin revealed that seven genetic variants within the ACOXL, CASP8, GRAMD1B, MYNN, PRKD2, TERC, and ZBTB7A|MAP2K2 loci were associated with TTFT at p < 0.05 level (Table 4). However, none of the associations with TTFT remained significant after correction for multiple testing, suggesting that these susceptibility variants for CLL do not have a relevant role in determining disease progression.
In line with these data, a meta-analysis of our data including data from a previous GWAS confirmed that none of these loci have a significant impact on TTFT (Table 5). These findings support the notion of a null effect of susceptibility variants on disease progression in CLL.
As expected, the association of weighted and unweighted PRSs built with those variants associated with TTFT in the CRuCIAL cohort was very modest (HRUnweighted = 1.26, p = 6.20 × 10−4 and HRWeighted = 1.32, p = 5.17 × 10−4 and HRUnweighted_Scaled_80% = 1.29, p = 4.40 × 10−5 and HRWeighted_Scaled_80% = 1.34, p = 6.60 × 10−5). Therefore, we were able to confirm that these PRSs were not useful in accurately predicting disease progression (AUROCUnweighted = 0.59, AUROCWeighted = 0.60, AUROCUnweighted_Scaled_80% = 0.61 and AUROCWeighted_Scaled_80% = 0.61; Table 6).

3. Discussion

This is the largest study evaluating the association of GWAS-identified susceptibility variants for CLL with OS, and one of the first studies assessing the effect of GWAS-identified susceptibility variants for CLL in disease progression. Even though we found potentially interesting associations between ten SNPs within the CAMK2D, CASP8, CFLAR, CXXC1, GPR37, IRF8, LEF1, MYNN, PRKD2, and TERC loci and the OS of CLL patients, none of these associations remained significant after correction for multiple testing. As expected, we found a modest association between weighted and unweighted PRSs and OS, which increased the prediction capacity by only 7%. These findings suggest that susceptibility variants for CLL do not have a large influence on OS, which is in agreement with a previous study that, using a similar approach, demonstrated that susceptibility variants do not influence the OS of patients diagnosed with multiple myeloma, another B cell malignancy [13].
This study failed to find a statistically significant association between GWAS-identified risk variants for CLL and TTFT. We found that only seven SNPs within the ACOXL, CASP8, GRAMD1B, MYNN, PRKD2, TERC, and ZBTB7A|MAP2K2 loci showed an association with TTFT at p < 0.05 level. None of these associations remained significant after correction for multiple testing, and a meta-analysis of our data with those from a previous GWAS on TTFT confirmed the null effect of these variants on disease progression. In agreement with these results, we found that weighted and unweighted PRSs did not have the capacity to predict TTFT. Nonetheless, in light of the relatively low power of our study (80% of power to detect an HR of 1.45 for a SNP with a frequency of 0.25) and the sparse number of studies assessing the impact of GWAS-identified risk variants for CLL on OS and TTFT, we cannot rule out the possibility that some of these SNPs might have a stronger effect on the modulation of OS and disease progression. In fact, we found it interesting that carriers of the CXXC1rs1036935A allele had poorer OS, as our team has previously reported that the presence of this allele is associated with decreased numbers of CD19+CD20+ B cells [14], a subtype of cells poorly expressed in CLL patients. The CXXC1 locus encodes for a protein of the SETD1 complex, which acts as an epigenetic transcriptional activator; if deregulated, it can lead to tumor progression and poorer survival [15].
This study has both strengths and weaknesses. The major strengths of this study are the large number of genetic markers analyzed and the relatively large size of the study population. Another strength is the comprehensive functional analysis conducted in the HFGP cohort, which included cQTL data after stimulation of whole blood, PBMCs, and MDMs with LPS, PHA, Pam3Cys, CpG, and B. burgdorferi and E. coli, as well as data on serological and plasmatic inflammatory proteins, serum steroid hormones, and blood-derived immune cell types. A limitation of this study is its multicentric nature, with inevitable drawbacks such as the impossibility of uniformly collecting medication history and Rai–Binet status for a significant proportion of the patients analyzed. In addition, considering that all study participants included in this study were of European ancestry, we could not determine the impact of GWAS-identified variants on patient survival and TTFT in other ethnic or ancestral populations.

4. Materials and Methods

4.1. Study Participants

This study included 1039 CLL patients ascertained through the CRuCIAL consortium. CLL patients were diagnosed following the updated international criteria [5]. Study participants were of European ancestry, and provided their written informed consent to participate in the study, which was approved by the ethical review committee of participant institutions: Virgen de las Nieves University Hospital (Granada, Spain, 0760-N-18); University Hospital of Salamanca (Salamanca, Spain, PI90/07/2018); Hospital del Mar (Barcelona, Spain); Catalan Institute of Oncology (Barcelona, Spain); Morales Meseguer University Hospital (Murcia, Spain); Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP) group (Spain); University of Modena and Reggio Emilia, AOU Policlinico (Modena, Italy); University of Pisa (Pisa, Italy), Wroclaw Medical University (Wroclaw, Poland); and the Radboud University Medical Center (Nijmegen, The Netherlands, 2011/299). A detailed description of the study cohort has been reported elsewhere [14]. The main characteristics of the patients are included in Table 1. This study followed the Declaration of Helsinki.

4.2. SNP Selection and Genotyping

A total of 41 single nucleotide polymorphisms (SNPs) were selected based on published GWAS, functionality data, and linkage disequilibrium between the SNPs (Supplementary Table S1) [14]. Genotyping of selected SNPs was carried out at GENYO (Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain) using KASPar® assays (LGC Genomics, Hoddesdon, UK) according to previously reported protocols. For internal quality control, ~5% of samples were randomly selected and included as duplicates. Concordance between the original and the duplicate samples for the selected SNPs was ≥99.0%. Call rate was higher than 90%.

4.3. Statistical Analysis and Meta-Analysis

The Hardy–Weinberg Equilibrium (HWE) test was performed in the control group using a standard observed–expected chi-square (χ2) test. The primary outcome was OS and the endpoint was defined as death from any cause. Survival time was calculated as the time from CLL diagnosis until the occurrence of the study endpoint, censoring at the date of death or the last observed follow-up time. The second outcome was time to first treatment (TTFT), defined as the interval between CLL diagnosis and date of the first treatment or last follow-up, while the endpoint was defined as death from any cause. Association with OS and TTFT, defined as hazard ratio (HR), was calculated for each SNP using Cox regression analysis adjusted for age, sex, and country of origin. Considering the number of SNPs and the number of inheritance models tested (log-additive, dominant, and recessive), we set a significance threshold to 0.00041 (0.05/41/3) using the Bonferroni correction. Association analyses were performed using STATA (v12.1; Stata Corp, College Station, TX, USA) and power calculations were estimated using the survSNP package in R (v4.1.1; R Core Team, 2018).
In order to confirm potentially interesting associations with disease progression, we conducted a meta-analysis of the CRuCIAL data with those from a previous GWAS [12] using METAL and assuming a fixed-effect model; the I2 statistic was used to assess statistical heterogeneity between cohorts. Next, in order to confirm whether susceptibility variants could predict OS and disease progression, we computed weighted and unweighted polygenic risk scores (PRSs) considering those SNPs associated with OS and TTFT at a threshold of p < 0.05. We built PRSs considering either subjects with a call rate of 100% (n = 891 and 290) or 80% (n = 1003 and 323) for OS and TTFT, respectively. PRS is an approach that estimates the individual risk to a phenotype or disease, which is calculated as a sum of their genotypes weighted by corresponding genotype effect sizes from summary statistic data. A detailed explanation of how the PRS scores were generated has been published in [16].

4.4. Functional Effect of the GWAS-Identified Risk Variants on Immune Responses

Mechanistically, we investigated whether selected SNPs were correlated with production of nine cytokines after in vitro stimulation of peripheral mononuclear cells (PBMCs) from 408 healthy subjects from the Human Functional Genomic Project (HFGP) cohort with LPS (100 ng/mL, Sigma-Aldrich, St. Louis, MO, USA), PHA (10 μg/mL, Sigma-Aldrich, St. Louis, MO, USA), Pam3Cys (10 μg/mL, EMC microcollections, Tuebingen, Germany), CpG (100 ng/mL, InvivoGen, San Diego, CA, USA), and B. burgdorferi (ATCC strain 35210) and E. coli (ATCC 25922). In addition, we investigated the correlation between SNPs and circulating concentrations of 103 serum and plasmatic inflammatory proteins, absolute numbers of 91 blood-derived immune cell populations (Supplementary Tables S2 and S3) and 7 plasma steroid hormones. Experimental protocols for the functional experiments have been previously described in detail [17,18]. Functional results for selected SNPs have been previously published by our team as part of a study in the context of the CRuCIAL consortium that aimed at validating the associations of GWAS-identified risk variants for CLL [14].

5. Conclusions

This study suggests that susceptibility variants for CLL do not substantially impact the overall survival of CLL patients, and confirms previous results suggesting the null effect of these variants on TTFT.

Supplementary Materials

The supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24098005/s1.

Author Contributions

J.S. designed and coordinated the study. A.J.C.-S., J.M.S.-M. and P.G.-M. were involved in the generation of genetic data from the CRuCIAL cohort. A.J.C.-S. and J.S. performed genetic association analyses, computed PRSs and drafted the manuscript. J.S. obtained funding and performed data quality control. M.G.N., R.t.H. and Y.L. provided the functional raw data from the HFGP cohort and J.S. performed the statistical analysis of functional data. A.M., Y.B., S.L., A.C.-G., Y.N., B.E., J.J.R.-S., E.M.P., R.M. (Rossana Maffei), G.B., M.G., J.R.C., R.M. (Roberto Marasca), M.Á.L.-N., T.C.-L., H.T., I.G., D.C. (Daniele Campa), V.M., S.d.S., R.M.-G., M.G.-Á., T.D.-S., A.J., A.B., A.D.N., M.L., S.L.S., K.H., Y.L., S.I.B., D.C. (Delphine Casabonne), M.A., A.P., M.G.N., A.F. and F.C. provided samples and data from CLL patients. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the European Union’s Horizon 2020 research and innovation program, N° 856620 and by grants from the Instituto de Salud Carlos III and FEDER (Madrid, Spain; PI17/02256 and PI20/01845) and from the Consejería de Transformación Económica, Industria, Conocimiento y Universidades y FEDER (PY20/01282). “The Mayo studies in InterLymph were supported in part by the US National Cancer Institute grants P50 CA97274 and R01 CA92153.”

Institutional Review Board Statement

This study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Boards of all institutions participating in the recruitment of CLL patients and/or healthy donors (Virgen de las Nieves University Hospital (Granada, Spain, 0760-N-18); University Hospital of Salamanca (Salamanca, Spain, PI90/07/2018); Hospital del Mar (Barcelona, Spain); Catalan Institute of Oncology (Barcelona, Spain); Morales Meseguer University Hospital (Murcia, Spain); Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP) group (Spain); University of Modena and Reggio Emilia, AOU Policlinico (Modena, Italy); University of Pisa (Pisa, Italy), Wroclaw Medical University (Wroclaw, Poland) and by the Radboud University Medical Center (Nijmegen, The Netherlands, 2011/299)).

Informed Consent Statement

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

Data Availability Statement

Genetic data from the CRuCIAL population can be accessed at https://ftp.genyo.es/ (accessed on 28 February 2023) and are available upon reasonable request. Functional data used in this study are available at BBMRI-NL data infrastructure (https://hfgp.bbmri.nl/, accessed on 28 February 2023), where they have been meticulously catalogued and archived using the MOLGENIS open-source platform for scientific data. This allows flexible data querying and download, including sufficiently rich metadata and interfaces for machine processing (R statistics, REST API) using FAIR principles to optimize Findability, Accessibility, Interoperability, and Reusability. These datasets are not publicly available because they contain information that could compromise the research participants’ privacy.

Acknowledgments

We kindly thank all individuals who agreed to participate in the study, as well as all cooperating physicians and students.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Alaggio, R.; Amador, C.; Anagnostopoulos, I.; Attygalle, A.D.; Araujo, I.B.O.; Berti, E.; Bhagat, G.; Borges, A.M.; Boyer, D.; Calaminici, M.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 2022, 36, 1720–1748. [Google Scholar] [CrossRef] [PubMed]
  2. Yao, Y.; Lin, X.; Li, F.; Jin, J.; Wang, H. The global burden and attributable risk factors of chronic lymphocytic leukemia in 204 countries and territories from 1990 to 2019: Analysis based on the global burden of disease study 2019. Biomed. Eng. Online 2022, 21, 4. [Google Scholar] [CrossRef] [PubMed]
  3. Parikh, S.A.; Rabe, K.G.; Kay, N.E.; Call, T.G.; Ding, W.; Schwager, S.M.; Bowen, D.A.; Conte, M.; Jelinek, D.F.; Slager, S.L.; et al. Chronic lymphocytic leukemia in young (≤55 years) patients: A comprehensive analysis of prognostic factors and outcomes. Haematologica 2014, 99, 140–147. [Google Scholar] [CrossRef] [PubMed]
  4. Alrawashdh, N.; Sweasy, J.; Erstad, B.; McBride, A.; Persky, D.O.; Abraham, I. Survival trends in chronic lymphocytic leukemia across treatment eras: US SEER database analysis (1985–2017). Ann. Hematol. 2021, 100, 2501–2512. [Google Scholar] [CrossRef] [PubMed]
  5. Hallek, M.; Al-Sawaf, O. Chronic lymphocytic leukemia: 2022 update on diagnostic and therapeutic procedures. Am. J. Hematol. 2021, 96, 1679–1705. [Google Scholar] [CrossRef] [PubMed]
  6. Rossi, D.; Rasi, S.; Spina, V.; Bruscaggin, A.; Monti, S.; Ciardullo, C.; Deambrogi, C.; Khiabanian, H.; Serra, R.; Bertoni, F.; et al. Integrated mutational and cytogenetic analysis identifies new prognostic subgroups in chronic lymphocytic leukemia. Blood 2013, 121, 1403–1412. [Google Scholar] [CrossRef] [PubMed]
  7. Berndt, S.I.; Camp, N.J.; Skibola, C.F.; Vijai, J.; Wang, Z.; Gu, J.; Nieters, A.; Kelly, R.S.; Smedby, K.E.; Monnereau, A.; et al. Meta-analysis of genome-wide association studies discovers multiple loci for chronic lymphocytic leukemia. Nat. Commun. 2016, 7, 10933. [Google Scholar] [CrossRef] [PubMed]
  8. Law, P.J.; Berndt, S.I.; Speedy, H.E.; Camp, N.J.; Sava, G.P.; Skibola, C.F.; Holroyd, A.; Joseph, V.; Sunter, N.J.; Nieters, A.; et al. Genome-wide association analysis implicates dysregulation of immunity genes in chronic lymphocytic leukaemia. Nat. Commun. 2017, 8, 14175. [Google Scholar] [CrossRef] [PubMed]
  9. Slager, S.L.; Rabe, K.G.; Achenbach, S.J.; Vachon, C.M.; Goldin, L.R.; Strom, S.S.; Lanasa, M.C.; Spector, L.G.; Rassenti, L.Z.; Leis, J.F.; et al. Genome-wide association study identifies a novel susceptibility locus at 6p21.3 among familial CLL. Blood 2011, 117, 1911–1916. [Google Scholar] [CrossRef] [PubMed]
  10. Di Bernardo, M.C.; Crowther-Swanepoel, D.; Broderick, P.; Webb, E.; Sellick, G.; Wild, R.; Sullivan, K.; Vijayakrishnan, J.; Wang, Y.; Pittman, A.M.; et al. A genome-wide association study identifies six susceptibility loci for chronic lymphocytic leukemia. Nat. Genet. 2008, 40, 1204–1210. [Google Scholar] [CrossRef] [PubMed]
  11. Call, T.G.; Phyliky, R.L.; Noel, P.; Habermann, T.M.; Beard, C.M.; O’Fallon, W.M.; Kurland, L.T. Incidence of chronic lymphocytic leukemia in Olmsted County, Minnesota, 1935 through 1989, with emphasis on changes in initial stage at diagnosis. Mayo Clin. Proc. 1994, 69, 323–328. [Google Scholar] [CrossRef] [PubMed]
  12. Lin, W.Y.; Fordham, S.E.; Sunter, N.; Elstob, C.; Rahman, T.; Willmore, E.; Shepherd, C.; Strathdee, G.; Mainou-Fowler, T.; Piddock, R.; et al. Genome-wide association study identifies risk loci for progressive chronic lymphocytic leukemia. Nat. Commun. 2021, 12, 665. [Google Scholar] [CrossRef] [PubMed]
  13. Macauda, A.; Clay-Gilmour, A.; Hielscher, T.; Hildebrandt, M.A.T.; Kruszewski, M.; Orlowski, R.Z.; Kumar, S.K.; Ziv, E.; Orciuolo, E.; Brown, E.E.; et al. Does a Multiple Myeloma Polygenic Risk Score Predict Overall Survival of Patients with Myeloma? Cancer Epidemiol. Biomark. Prev. 2022, 31, 1863–1866. [Google Scholar] [CrossRef] [PubMed]
  14. Garcia-Martin, P.; Diez, A.M.; Maldonado, J.M.S.; Serrano, A.J.C.; Ter Horst, R.; Benavente, Y.; Landi, S.; Macauda, A.; Clay-Gilmour, A.; Hernandez-Mohedo, F.; et al. Validation and functional characterization of GWAS-identified variants for chronic lymphocytic leukemia: A CRuCIAL study. Blood Cancer J. 2022, 12, 79. [Google Scholar] [CrossRef] [PubMed]
  15. Wu, M.; Fan, B.; Guo, Q.; Li, Y.; Chen, R.; Lv, N.; Diao, Y.; Luo, Y. Knockdown of SETDB1 inhibits breast cancer progression by miR-381-3p-related regulation. Biol. Res. 2018, 51, 39. [Google Scholar] [CrossRef] [PubMed]
  16. Canzian, F.; Piredda, C.; Macauda, A.; Zawirska, D.; Andersen, N.F.; Nagler, A.; Zaucha, J.M.; Mazur, G.; Dumontet, C.; Watek, M.; et al. A polygenic risk score for multiple myeloma risk prediction. Eur. J. Hum. Genet. 2022, 30, 474–479. [Google Scholar] [CrossRef] [PubMed]
  17. Li, Y.; Oosting, M.; Smeekens, S.P.; Jaeger, M.; Aguirre-Gamboa, R.; Le, K.T.T.; Deelen, P.; Ricano-Ponce, I.; Schoffelen, T.; Jansen, A.F.M.; et al. A Functional Genomics Approach to Understand Variation in Cytokine Production in Humans. Cell 2016, 167, 1099–1110.e14. [Google Scholar] [CrossRef] [PubMed]
  18. Orru, V.; Steri, M.; Sole, G.; Sidore, C.; Virdis, F.; Dei, M.; Lai, S.; Zoledziewska, M.; Busonero, F.; Mulas, A.; et al. Genetic variants regulating immune cell levels in health and disease. Cell 2013, 155, 242–256. [Google Scholar] [CrossRef] [PubMed]
Table 1. Patient characteristics of the CRuCIAL cohort used for OS and TTFT analyses.
Table 1. Patient characteristics of the CRuCIAL cohort used for OS and TTFT analyses.
CRuCIAL Cohort (1039 CLL Cases for OS Analysis)CRuCIAL Cohort (354 CLL Cases for TTFT Analysis)
 Age (years)65.87  ± 11.05 Age (years)65.03  ± 10.94
 Sex ratio (male/female)1.57 (636/403) Sex ratio (male/female)1.62 (219/135)
 Country of origin (alive/decesead)  Country of origin
    Spain754 (536/218)    Spain227 (174/73)
    Italy285 (216/69)     Italy127 (86/41)
 Median follow-up time (months) 76.77 (50–123) Median TTFT (days) 759.49 (31–1148.74)
    Alive76 (52.1–123)    Alive830.81 (44.13–1308.93)
    Deceased77 (44–123)     Deceased564.83 (13.10–800)
 Status at follow-up  Status at follow-up
    Alive752 (72.38)    Alive239 (68.29)
    Deceased287 (27.62)    Deceased111 (31.71)
 Binet Stage  Binet stage
    A647 (81.48)    A201 (64.84)
    B99 (12.46)    B68 (21.94)
    C48 (06.04)    C41 (13.23)
 Rai Stage  Rai stage
    0597 (65.67)    0124 (37.35)
    I152 (16.72)    I105 (31.63)
    II114 (12.57)    II63 (18.98)
    III12 (01.31)    III16 (04.82)
    IV34 (03.73)    IV24 (07.23)
Data are mean ± standard deviation, n (%), or percentiles (25th–75th percentiles).
Table 2. Association of GWAS-identified risk variants for CLL and OS.
Table 2. Association of GWAS-identified risk variants for CLL and OS.
SNPChr.Nearby GeneRisk AlleleHR (95%CI) δpHR (95%CI) ϮpHR (95%CI) ¥p
rs436825318AC107990.1||NFE2L3P1C0.94 (0.78–1.14)0.5390.93 (0.59–1.45)0.7460.93 (0.73–1.18)0.545
rs14392872ACOXLT1.03 (0.88–1.21)0.7000.99 (0.76–1.29)0.9571.10 (0.84–1.44)0.481
rs580556742ACOXLC1.08 (0.90–1.30)0.4241.12 (0.89–1.42)0.3351.02 (0.64–1.61)0.936
rs794400411ASCL2||C11orf21T0.97 (0.82–1.15)0.7351.06 (0.79–1.43)0.6990.89 (0.69–1.16)0.394
rs498785518BCL2G0.97 (0.71–1.32)0.8480.68 (0.10–4.87)0.7010.98 (0.71–1.34)0.886
rs265182311C11orf21|TSPAN32A0.95 (0.81–1.12)0.5231.03 (0.78–1.36)0.8570.85 (0.65–1.11)0.233
rs14765694CAMK2DG1.12 (0.95–1.32)0.1761.31 (1.03–1.67)0.0280.92 (0.64–1.32)0.643
rs37698252CASP8T1.20 (1.02–1.43)0.0331.23 (0.93–1.63)0.1501.32 (1.01–1.73)0.041
rs75589112CFLARA1.19 (1.01–1.41)0.0401.22 (0.91–1.63)0.1751.30 (1.00–1.67)0.046
rs103693518CXXC1A1.27 (1.07–1.51)0.0081.34 (1.06–1.70)0.0151.43 (0.98–2.10)0.066
rs13597429DMRTA1G1.05 (0.89–1.23)0.5751.02 (0.78–1.34)0.8751.11 (0.85–1.45)0.441
rs65461492DTNBG1.09 (0.91–1.31)0.3561.06 (0.84–1.35)0.6071.29 (0.86–1.93)0.222
rs98807723EOMES|LINC01980T0.98 (0.84–1.14)0.7871.04 (0.80–1.34)0.7870.90 (0.69–1.18)0.455
rs130157982FAM126BA1.06 (0.88–1.28)0.5481.00 (0.64–1.54)0.9891.10 (0.87–1.39)0.444
rs658616310FASA0.99 (0.84–1.17)0.9161.08 (0.80–1.45)0.6310.92 (0.71–1.20)0.554
rs22677087GPR37T0.86 (0.72–1.02)0.0810.91 (0.70–1.19)0.5040.70 (0.51–0.97)0.030
rs295319611GRAMD1BG0.85 (0.70–1.04)0.1230.70 (0.42–1.17)0.1760.85 (0.67–1.09)0.203
rs3592364311GRAMD1BG0.88 (0.72–1.07)0.1920.80 (0.63–1.02)0.0681.12 (0.71–1.78)0.631
rs38004616ILRUNC0.92 (0.67–1.27)0.6200.91 (0.65–1.27)0.5621.20 (0.30–4.86)0.795
rs93925046IRF4A0.88 (0.74–1.04)0.1430.80 (0.60–1.08)0.1450.88 (0.68–1.13)0.322
rs39185516IRF8A0.87 (0.74–1.03)0.1040.99 (0.72–1.36)0.9460.74 (0.58–0.96)0.021
rs8985184LEF1A1.12 (0.94–1.33)0.2151.50 (1.00–2.26)0.0491.04 (0.82–1.33)0.721
rs346762231MDS2C0.99 (0.83–1.18)0.8990.88 (0.61–1.27)0.4911.03 (0.81–1.30)0.803
rs572142774MYL12BP2||LINC02363T1.02 (0.86–1.21)0.8051.01 (0.79–1.29)0.9261.06 (0.77–1.46)0.729
rs109365993MYNNC0.91 (0.74–1.11)0.3440.57 (0.36–0.90)0.0160.99 (0.77–1.26)0.922
rs117156043NCK1T0.98 (0.80–1.20)0.8311.06 (0.61–1.85)0.8420.96 (0.74–1.23)0.723
rs648988212OAS3G1.09 (0.92–1.29)0.3091.16 (0.90–1.50)0.2401.06 (0.78–1.45)0.696
rs14052222ODF3BT1.02 (0.86–1.20)0.8541.11 (0.87–1.41)0.4100.87 (0.61–1.24)0.431
rs22362566OPRM1||IPCEF1C1.03 (0.87–1.21)0.7520.96 (0.74–1.23)0.7291.14 (0.86–1.51)0.348
rs1163756515PCAT29|LOC107984788G0.97 (0.82–1.16)0.7530.96 (0.74–1.25)0.7530.97 (0.71–1.33)0.842
rs172464047POT1C0.95 (0.79–1.14)0.5720.95 (0.61–1.47)0.8180.93 (0.74–1.18)0.557
rs25117148POU5F1P2||ODF1G0.91 (0.77–1.08)0.2680.92 (0.72–1.17)0.4850.82 (0.59–1.14)0.240
rs1108384619PRKD2A1.20 (0.99–1.45)0.0611.16 (0.92–1.47)0.2091.62 (1.06–2.50)0.027
rs8880962QPCT||RNU6-1116PA0.95 (0.81–1.13)0.5900.97 (0.76–1.24)0.8370.88 (0.63–1.23)0.454
rs412714731RHOUG0.96 (0.74–1.25)0.7701.46 (0.64–3.32)0.3660.88 (0.64–1.21)0.432
rs737187796SERPINB6A1.03 (0.78–1.37)0.8231.04 (0.77–1.40)0.8060.97 (0.24–3.94)0.970
rs126388623TERCA0.89 (0.73–1.08)0.2290.62 (0.40–0.97)0.0370.94 (0.74–1.19)0.602
rs77055265TERTA1.07 (0.90–1.27)0.4681.00 (0.78–1.28)0.9971.25 (0.91–1.70)0.163
rs6190498711TMPRSS5||DRD2T0.94 (0.71–1.23)0.6320.94 (0.70–1.28)0.7120.73 (0.23–2.30)0.588
rs9260706TSBP1-AS1A1.11 (0.92–1.33)0.2851.19 (0.77–1.82)0.4331.12 (0.88–1.42)0.343
rs725427219ZBTB7A|MAP2K2A0.82 (0.66–1.02)0.0780.78 (0.61–1.00)0.0510.92 (0.50–1.69)0.794
Abbreviations: SNP, single nucleotide polymorphism; HR, hazards ratio. Significant results in bold (p < 0.05). δ Cox regression analysis assuming a log-additive model of inheritance. Ϯ Cox regression analysis assuming a dominant model. ¥ Cox regression analysis assuming a recessive model.
Table 3. Associations between unweighted and weighted PRSs and OS.
Table 3. Associations between unweighted and weighted PRSs and OS.
Polygenic Risk Scores (n = 891)AUROC
QuintilesHR 95%CI apAUROC (95%CI)
Unweighted, subjects with 100% call rate11.00-
21.06 (0.70–1.60)0.787
31.67 (1.15–2.43)0.007
41.42 (0.99–2.03)0.053
52.36 (1.56–3.58)5.30 × 10−5
Continuous b1.20 (1.09–1.31)8.70 × 10−50.56 (0.52–0.60)
Weighted, subjects with 100% call rate11.00-
21.33 (0.85–2.09)0.206
32.05 (1.34–3.15)0.001
41.68 (1.08–2.59)0.020
52.50 (1.63–3.83)2.40 × 10−5
Continuous b1.22 (1.11–1.33)1.80 × 10−50.57 (0.53–0.61)
Polygenic Risk Scores (n = 1003)AUROC
QuintilesHR 95%CI apAUROC (95%CI)
Unweighted, subjects with 80% call rate11.00-
20.99 (0.66–1.46)0.948
31.48 (1.05–2.11)0.027
41.36 (0.98–1.90)0.066
52.08 (1.41–3.07)2.41 × 10−4
Continuous b1.17 (1.08–1.28)2.32 × 10−40.55 (0.51–0.59)
Weighted, subjects with 80% call rate11.00-
21.29 (0.85–1.95)0.224
31.78 (1.19–2.67)0.005
41.57 (1.05–2.35)0.028
52.19 (1.48–3.26)9.80 × 10−5
Continuous b1.19 (1.09–1.29)7.61 × 10−50.56 (0.52–0.60)
a HR, hazards ratio; CI, confidence interval. All analyses were adjusted for age, sex, and country of origin; b The unit for the analysis with the continuous variable was the increment of one quintile.
Table 4. Association of GWAS-identified risk variants for CLL and TTFT.
Table 4. Association of GWAS-identified risk variants for CLL and TTFT.
SNPChr.Nearby GeneRisk AlleleHR (95%CI) δpHR (95%CI) ϮpHR (95%CI) ¥p
rs436825318AC107990.1||NFE2L3P1C1.03 (0.74–1.42)0.8710.62 (0.31–1.25)0.1841.18 (0.80–1.74)0.413
rs14392872ACOXLT1.05 (0.81–1.37)0.7080.98 (0.65–1.50)0.9431.18 (0.76–1.83)0.469
rs580556742ACOXLC1.39 (1.02–1.90)0.0361.60 (1.08–2.38)0.0191.18 (0.51–2.73)0.696
rs794400411ASCL2||C11orf21T1.04 (0.78–1.39)0.7691.01 (0.61–1.67)0.9671.09 (0.71–1.68)0.684
rs498785518BCL2G0.75 (0.44–1.27)0.281NANA0.73 (0.43–1.25)0.256
rs265182311C11orf21|TSPAN32A1.12 (0.85–1.46)0.4181.13 (0.73–1.76)0.5911.20 (0.77–1.85)0.423
rs14765694CAMK2DG1.01 (0.76–1.35)0.9511.02 (0.69–1.51)0.9150.99 (0.54–1.82)0.971
rs37698252CASP8T1.41 (1.06–1.87)0.0171.56 (0.98–2.47)0.0591.58 (1.00–2.48)0.048
rs75589112CFLARA1.21 (0.93–1.58)0.1631.23 (0.78–1.95)0.3751.35 (0.89–2.05)0.155
rs103693518CXXC1A1.13 (0.83–1.53)0.4461.14 (0.77–1.69)0.5031.24 (0.59–2.61)0.568
rs13597429DMRTA1G0.98 (0.74–1.30)0.8840.85 (0.54–1.32)0.4681.11 (0.72–1.73)0.636
rs65461492DTNBG0.95 (0.69–1.31)0.7690.90 (0.61–1.33)0.5871.14 (0.55–2.36)0.723
rs98807723EOMES|LINC01980T0.96 (0.73–1.24)0.7350.96 (0.64–1.43)0.8310.91 (0.56–1.48)0.717
rs130157982FAM126BA0.92 (0.68–1.23)0.5670.88 (0.45–1.70)0.6950.90 (0.61–1.33)0.605
rs658616310FASA0.81 (0.62–1.07)0.1390.80 (0.51–1.26)0.3390.72 (0.45–1.13)0.150
rs22677087GPR37T0.76 (0.58–1.00)0.0520.69 (0.46–1.03)0.0690.71 (0.43–1.16)0.172
rs295319611GRAMD1BG0.82 (0.59–1.14)0.2400.85 (0.34–2.10)0.7240.76 (0.50–1.16)0.202
rs3592364311GRAMD1BG0.71 (0.52–0.98)0.0400.68 (0.46–1.02)0.0610.54 (0.23–1.28)0.164
rs38004616ILRUNC0.97 (0.61–1.54)0.8810.94 (0.55–1.60)0.8081.17 (0.27–5.08)0.832
rs93925046IRF4A0.94 (0.72–1.23)0.6481.07 (0.63–1.81)0.8000.83 (0.55–1.26)0.380
rs39185516IRF8A1.14 (0.87–1.49)0.3361.28 (0.77–2.14)0.3411.15 (0.77–1.72)0.503
rs8985184LEF1A1.16 (0.87–1.54)0.3101.58 (0.82–3.04)0.1721.09 (0.74–1.62)0.650
rs346762231MDS2C1.28 (0.96–1.71)0.0981.96 (0.94–4.07)0.0731.24 (0.84–1.82)0.280
rs572142774MYL12BP2||LINC02363T1.10 (0.85–1.43)0.4561.28 (0.86–1.91)0.2250.97 (0.58–1.61)0.897
rs109365993MYNNC1.03 (0.73–1.46)0.8660.42 (0.21–0.83)0.0131.30 (0.85–1.98)0.222
rs117156043NCK1T0.78 (0.55–1.10)0.1580.59 (0.24–1.48)0.2630.77 (0.51–1.17)0.225
rs648988212OAS3G1.01 (0.76–1.34)0.9291.06 (0.70–1.59)0.7970.96 (0.56–1.63)0.868
rs14052222ODF3BT0.88 (0.66–1.18)0.3910.86 (0.58–1.28)0.4520.81 (0.43–1.53)0.523
rs22362566OPRM1||IPCEF1C1.06 (0.79–1.41)0.7040.79 (0.52–1.21)0.2791.50 (0.97–2.33)0.070
rs1163756515PCAT29|LOC107984788G0.90 (0.68–1.20)0.4821.04 (0.67–1.61)0.8740.67 (0.38–1.16)0.155
rs172464047POT1C1.14 (0.83–1.56)0.4251.63 (0.70–3.79)0.2571.08 (0.72–1.61)0.702
rs25117148POU5F1P2||ODF1G0.97 (0.72–1.31)0.8380.93 (0.61–1.40)0.7211.03 (0.57–1.86)0.914
rs1108384619PRKD2A1.34 (1.00–1.80)0.0501.21 (0.82–1.78)0.3312.31 (1.31–4.08)0.004
rs8880962QPCT||RNU6-1116PA1.02 (0.76–1.35)0.9120.96 (0.64–1.44)0.8511.14 (0.66–1.96)0.631
rs412714731RHOUG0.76 (0.50–1.13)0.1760.85 (0.26–2.76)0.7860.69 (0.42–1.13)0.139
rs737187796SERPINB6A1.15 (0.74–1.78)0.5361.09 (0.68–1.74)0.7342.86 (0.69–11.9)0.148
rs126388623TERCA0.94 (0.66–1.33)0.7150.40 (0.19–0.84)0.0151.12 (0.74–1.69)0.601
rs77055265TERTA0.92 (0.70–1.23)0.5890.79 (0.53–1.17)0.2401.13 (0.69–1.86)0.633
rs6190498711TMPRSS5||DRD2T1.28 (0.86–1.92)0.2281.35 (0.85–2.16)0.2031.23 (0.30–5.10)0.776
rs9260706TSBP1-AS1A1.00 (0.74–1.35)0.9870.87 (0.45–1.69)0.6841.04 (0.71–1.54)0.835
rs725427219ZBTB7A|MAP2K2A0.74 (0.51–1.07)0.1100.62 (0.41–0.96)0.0301.34 (0.58–3.09)0.497
Abbreviations: SNP, single nucleotide polymorphism; HR, hazards ratio. Significant results in bold (p < 0.05). δ Cox regression analysis assuming a log-additive model of inheritance. Ϯ Cox regression analysis assuming a dominant model of inheritance. ¥ Cox regression analysis assuming a recessive model of inheritance.
Table 5. Meta-analysis of association estimates of GWAS-identified risk variants for CLL and disease progression in the CRuCIAL cohorts with a previous GWAS.
Table 5. Meta-analysis of association estimates of GWAS-identified risk variants for CLL and disease progression in the CRuCIAL cohorts with a previous GWAS.
SNPChr.Nearby GeneRisk AlleleCRuCIAL Consortium
(354 CLL Cases)
Lin et al. (2021) [12]
(755 CLL Cases)
Meta-Analysis
(1109 CLL Cases)
HR (95%CI) δpHR (95%CI) δpHR (95%CI) δpphet
rs436825318AC107990.1||NFE2L3P1C1.03 (0.74–1.42)0.8711.03 (0.87–1.17)0.6841.03 (0.85–1.19)0.7260.985
rs14392872ACOXLT1.05 (0.81–1.37)0.7081.00 (0.86–1.12)0.9901.01 (1.16–0.88)0.8580.765
rs580556742ACOXLC1.39 (1.02–1.90)0.0360.98 (0.84–1.15)0.8371.08 (0.89–1.23)0.3980.096
rs794400411ASCL2||C11orf21T1.04 (0.78–1.39)0.769--1.04 (1.35–0.80)0.7831.000
rs498785518BCL2G0.75 (0.44–1.27)0.2810.82 (0.45–1.10)0.2300.78 (0.33–1.10)0.2050.734
rs265182311C11orf21|TSPAN32A1.12 (0.85–1.46)0.4180.93 (0.80–1.07)0.3020.98 (1.14–0.84)0.7960.303
rs14765694CAMK2DG1.01 (0.76–1.35)0.9510.99 (0.85–1.15)0.8981.00 (0.83–1.14)0.9550.917
rs37698252CASP8T1.41 (1.06–1.87)0.0171.10 (0.96–1.22)0.1491.01 (1.18–0.87)0.8700.018
rs75589112CFLARA1.21 (0.93–1.58)0.1630.81 (0.63–0.97)0.0190.94 (1.10–0.80)0.4210.048
rs103693518CXXC1A1.13 (0.83–1.53)0.4460.96 (0.83–1.13)0.6601.01 (1.19–0.85)0.9490.444
rs13597429DMRTA1G0.98 (0.74–1.30)0.8840.93 (0.76–1.07)0.3440.94 (0.77–1.10)0.5060.778
rs65461492DTNBG0.95 (0.69–1.31)0.7690.86 (0.73–1.01)0.0670.87 (0.64–1.06)0.1920.652
rs98807723EOMES|LINC01980T0.96 (0.73–1.24)0.7351.00 (0.87–1.12)0.9540.98 (1.13–0.86)0.8280.815
rs130157982FAM126BA0.92 (0.68–1.23)0.5670.78 (0.57–0.96)0.0150.85 (1.02–0.71)0.0760.556
rs658616310FASA0.81 (0.62–1.07)0.1391.00 (0.86–1.13)0.9720.95 (1.11–0.82)0.5270.249
rs22677087GPR37T0.76 (0.58–1.00)0.0520.98 (0.85–1.12)0.7430.91 (1.07–0.78)0.2730.176
rs295319611GRAMD1BG0.82 (0.59–1.14)0.2401.16 (0.99–1.31)0.0651.06 (0.84–1.23)0.5960.102
rs3592364311GRAMD1BG0.71 (0.52–0.98)0.0401.18 (1.00–1.39)0.0491.03 (0.84–1.19)0.7600.021
rs38004616ILRUNC0.97 (0.61–1.54)0.8811.19 (0.96–1.47)0.1051.12 (1.41–0.88)0.3630.426
rs93925046IRF4A0.94 (0.72–1.23)0.6481.03 (0.88–1.16)0.6781.00 (1.16–0.87)0.9700.597
rs39185516IRF8A1.14 (0.87–1.49)0.3361.00 (0.85–1.13)0.9671.04 (1.21–0.89)0.6240.477
rs8985184LEF1A1.16 (0.87–1.54)0.3100.92 (0.76–1.06)0.2820.98 (1.14–0.84)0.8180.234
rs346762231MDS2C1.28 (0.96–1.71)0.0981.06 (0.90–1.19)0.4671.11 (0.93–1.25)0.2100.347
rs572142774MYL12BP2||LINC02363T1.10 (0.85–1.43)0.4560.92 (0.79–1.06)0.2280.97 (1.13–0.84)0.7280.286
rs109365993MYNNC1.03 (0.73–1.46)0.8660.91 (0.70–1.08)0.3140.94 (0.72–1.13)0.5860.587
rs117156043NCK1T0.78 (0.55–1.10)0.1581.05 (0.87–1.27) η0.6140.96 (0.73–1.15)0.7190.207
rs648988212OAS3G1.01 (0.76–1.34)0.9291.10 (0.95–1.27)0.2131.06 (0.91–1.18)0.4500.588
rs14052222ODF3BT0.88 (0.66–1.18)0.3911.04 (0.90–1.20)0.6410.99 (1.16–0.85)0.9270.397
rs22362566OPRM1||IPCEF1C1.06 (0.79–1.41)0.7041.04 (0.91–1.20)0.5681.04 (0.88–1.18)0.5800.930
rs1163756515PCAT29|LOC107984788G0.90 (0.68–1.20)0.4820.89 (0.77–1.03)0.1120.88 (0.68–1.05)0.1820.958
rs172464047POT1C1.14 (0.83–1.56)0.4251.01 (0.85–1.15)0.8721.04 (0.86–1.19)0.6360.577
rs25117148POU5F1P2||ODF1G0.97 (0.72–1.31)0.8381.00 (0.87–1.14)0.9620.99 (0.83–1.13)0.8950.883
rs1108384619PRKD2A1.34 (1.00–1.80)0.0501.11 (0.94–1.30)0.2281.17 (1.41–0.98)0.0880.342
rs8880962QPCT||RNU6-1116PA1.02 (0.76–1.35)0.9121.08 (0.93–1.24)0.3241.06 (1.26–0.90)0.4840.801
rs412714731RHOUG0.76 (0.50–1.13)0.1760.92 (0.71–1.09)0.3680.87 (0.61–1.08)0.2470.453
rs737187796SERPINB6A1.15 (0.74–1.78)0.5360.74 (0.42–0.99)0.0401.23 (1.59–0.95)0.1130.750
rs126388623TERCA0.94 (0.66–1.33)0.7150.95 (0.76–1.11)0.5670.95 (1.14–0.79)0.5820.958
rs77055265TERTA0.92 (0.70–1.23)0.589--0.92 (1.27–0.67)0.6211.000
rs6190498711TMPRSS5||DRD2T1.28 (0.86–1.92)0.2281.06 (0.87–1.29)0.5701.11 (1.40–0.89)0.3570.481
rs9260706TSBP1-AS1A1.00 (0.74–1.35)0.9871.07 (0.91–1.19)0.3821.05 (1.25–0.89)0.5700.742
rs725427219ZBTB7A|MAP2K2A0.74 (0.51–1.07)0.1101.12 (0.93–1.35)0.2421.00 (1.22–0.82)0.9910.096
Abbreviations: SNP, single nucleotide polymorphism; HR, hazards ratio. CI, confidence interval; Meta-analysis was performed assuming a fixed-effect model. Significant results in bold (p < 0.05). η Authors report the effect found for the rs62410363 (a SNP in strong linkage disequilibrium with the rs11715604, r2 = 0.97). δ Cox regression analyses were adjusted for age, sex, and country of origin and were calculated according to log-additive model of inheritance.
Table 6. Associations between unweighted and weighted PRSs and disease progression.
Table 6. Associations between unweighted and weighted PRSs and disease progression.
Polygenic Risk Scores (n = 290)AUROC
QuintilesHR 95%CI apAUROC (95%CI)
Unweighted, subjects with 100% call rate11.00-
21.23 (0.68–2.22)0.487
3--
41.89 (1.08–3.31)0.026
52.66 (1.45–4.88)1.50 × 10−3
Continuous b1.26 (1.11–1.45)6.20 × 10−40.59 (0.52–0.66)
Weighted, subjects with 100% call rate11.00-
22.22 (1.05–4.71)0.037
31.45 (0.66–3.16)0.353
42.34 (1.14–4.79)0.020
53.87 (1.89–7.94)2.10 × 10−4
Continuous b1.32 (1.13–1.54)5.17 × 10−40.60 (0.53–0.67)
Polygenic risk scores (n = 323)AUROC
QuintilesHR 95%CI apAUROC (95%CI)
Unweighted, subjects with 80% call rate11.00
21.27 (0.73–2.21)0.392
3--
41.85 (1.07–3.19)0.027
53.00 (1.75–5.12)5.90 × 10−5
Continuous b1.29 (1.14–1.46)4.40 × 10−50.61 (0.54–0.67)
Weighted, subjects with 80% call rate11.00
21.89 (0.93–3.85)0.080
31.64 (0.81–3.32)0.172
42.64 (1.37–5.10)3.80 × 10−3
53.58 (1.85–6.93)1.50 × 10−4
Continuous b1.34 (1.16–1.54)6.60 × 10−50.61 (0.55–0.67)
a HR, hazards ratio; CI, confidence interval. All analyses were adjusted for age, sex, and geographic region of origin. b The unit for the analysis with the continuous variable was the increment of one quintile.
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

Cabrera-Serrano, A.J.; Sánchez-Maldonado, J.M.; ter Horst, R.; Macauda, A.; García-Martín, P.; Benavente, Y.; Landi, S.; Clay-Gilmour, A.; Niazi, Y.; Espinet, B.; et al. Do GWAS-Identified Risk Variants for Chronic Lymphocytic Leukemia Influence Overall Patient Survival and Disease Progression? Int. J. Mol. Sci. 2023, 24, 8005. https://doi.org/10.3390/ijms24098005

AMA Style

Cabrera-Serrano AJ, Sánchez-Maldonado JM, ter Horst R, Macauda A, García-Martín P, Benavente Y, Landi S, Clay-Gilmour A, Niazi Y, Espinet B, et al. Do GWAS-Identified Risk Variants for Chronic Lymphocytic Leukemia Influence Overall Patient Survival and Disease Progression? International Journal of Molecular Sciences. 2023; 24(9):8005. https://doi.org/10.3390/ijms24098005

Chicago/Turabian Style

Cabrera-Serrano, Antonio José, José Manuel Sánchez-Maldonado, Rob ter Horst, Angelica Macauda, Paloma García-Martín, Yolanda Benavente, Stefano Landi, Alyssa Clay-Gilmour, Yasmeen Niazi, Blanca Espinet, and et al. 2023. "Do GWAS-Identified Risk Variants for Chronic Lymphocytic Leukemia Influence Overall Patient Survival and Disease Progression?" International Journal of Molecular Sciences 24, no. 9: 8005. https://doi.org/10.3390/ijms24098005

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