Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) refers to the expansion of cells of hematopoietic lineage that carry acquired somatic alterations associated with hematologic malignancies. The most commonly altered genes giving rise to CHIP are DNMT3A, TET2, and ASXL1. However, advanced sequencing technologies have resulted in highly sensitive detection of clonal hematopoiesis beyond these known driver genes. In practice, CHIP is commonly identified as an incidental finding in liquid and tissue biopsies of solid tumor patients. CHIP can have broad clinical consequences, given its association with hematologic malignancies and non-malignant diseases. CHIP can also interfere with next-generation DNA sequencing results, so clinicians should pay careful attention when these results are being used to guide therapy. Future research is needed to determine how solid tumor malignancies and their treatments alter the progression of CHIP, and in turn, how CHIP might be used to improve treatment selection and outcomes for patients with solid tumors.
Introduction
There are approximately one trillion blood cells arising daily from hematopoietic stem cells (HSC) in an adult human body.(1) This is sustained by an estimated 50 to 200 thousand HSC.(2) Somatic nucleotide alterations occur at approximately 1.14 mutations per cell division in cells of the hematopoietic lineage.(2) Over a lifespan, while most cells acquire somatic mutations that do not affect function, some will acquire mutations that may improve fitness and result in selective expansion of these cells without corresponding clinical manifestations.(3) This is called clonal hematopoiesis (CH). The term CH is sometimes used interchangeably with the term clonal hematopoiesis of indeterminate potential (CHIP). However, CHIP in general has a narrower definition, referring to clonal hematopoiesis in individuals without evidence of hematologic malignancies but with mutations in genes associated with hematologic malignancies, and detected at >2% variant allele frequency (VAF).(4) CHIP is associated with aging and, while it is exceedingly rare in people under the age of 40, the incidence increases with every decade of life reaching a prevalence of approximately 10 to 20% among individuals over 70 years.(5–7) However, deeper sequencing with error correction allowing for more sensitive detection of CH (i.e., smaller clones with lower VAF), does result in more frequent detection of CH in younger individuals.(8,9)
In the general population, somatic alterations in the genes associated with hematologic malignancies which gives rise to CHIP occur as the result of one of four processes known to contribute to age-associated mutagenesis: deamination of 5-methylcytosine to thymine, nucleotide insertions and deletions after double strand break repair, polymerase errors during DNA replication, and chromosomal rearrangements.(10) The mechanism of enhanced fitness likely varies based on the specific mutation and biological context.(8,11) While both driver and passenger genes can be used to detect CH, results from a number of studies suggest that most CH events, especially those detected by the more sensitive sequencing methods, may not be explained by known driver mutations. (7,12–15) Nevertheless, known cancer-associated driver genes have been more extensively evaluated in the context of CHIP. For example, mutations in DNMT3A, TET2, ASXL1, TP53, JAK2, PPM1D, ATM, CBL, SF3B1, BCORL1, GNAS, and CHEK2 are some of the most common CHIP-defining genes and constitute over 90% of all CHIP alterations (Table 1).(4,16)
Table 1.
List of 10 most common genes altered in clonal hematopoiesis of indeterminate potential (CHIP) and their function
Gene | Full Name | Function (from ncbi.nlm.nih.gov) |
---|---|---|
DNMT3A | DNA methyltransferase 3; | de novo methylation, epigenetic regulation |
TET2 | TET methylcytosine dioxygenase 2 | demethylation, epigenetic regulation |
ASXL1 | ASXL transcriptional regulator 1 | Chromatin binding protein |
PPM1D | Protein phosphatase, Mg2+/Mn2+ dependent 1D | Suppresses p53-mediated transcription and apoptosis |
TP53 | Tumor protein p53 | Tumor suppressor |
CHEK2 | Checkpoint kinase 2 | DNA damage response and tumor suppressor |
ATM | ATM serine/threonine kinase | Cell cycle checkpoint, DNA damage response |
SF3B1 | Splicing factor 3b subunit 1 | Splicing machinery |
JAK2 | Janus kinase 2 | Tyrosine kinase central to cytokine and growth factor signaling |
CBL | Cbl proto-oncogene | Proto-oncogene, cell signaling |
In this narrative, we summarize the prevalence and risk factors for CHIP in patients with solid tumor malignancies, review the clinical implications of CHIP among patients with cancer, and discuss ways in which CHIP might serve as a biomarker in the future for prognosis and therapeutic decision making (Figure 1).
Figure 1.
Risk factors for the development of clonal hematopoiesis and implications among individuals with solid tumor malignancies.
Risk factors
A number of risk factors for the development of CHIP have been identified. Some, but not all, demographic factors have been identified as risk factors for CHIP. Chronologic age has consistently shown to be a risk factor for CHIP, having been identified across many studies.(6,7,12,17–29) Further supporting a biologic basis associating CHIP with age, a study of 5,522 people showed that CHIP was also associated with biologic age as measured by epigenetic changes.(30) Among 19,606 women age 40–70, premature menopause was independently associated with CHIP with 36% increased odds of having CHIP, especially with DNMT3A mutations (95% 1.10–1.79; P=0.007) raising the possibility that hormonal levels, and particularly estrogen, may play a role in the development of CHIP.(31) Male sex and White race/Non-Hispanic ethnicity have also been implicated as risk factors for the development of CHIP in some, but not all studies. (16,19,20,27–29)
There are conflicting data as to whether or not there are genetic predispositions to CHIP. Two twin studies, one with 299 pairs of twins ages 73 to 94, and another with 79 twin pairs age 70–99, did not find any evidence of inherited predisposition to CHIP.(32,33) In a separate study, a germline deletion of intron 3 in the TERT gene (telomerase reverse transcriptase gene) was associated with a 37% increased risk of CHIP in a population of 262 subjects from Iceland.(12) This was replicated in a larger study of 97,631 peripheral blood whole genome sequencing study from the US-based TOPMed program.(28) Mechanistically, a reciprocal relationship between CHIP and telomerase activity was reported.(34) In addition to the association with TERT, two additional gene areas of interest were identified –an intergenic region near TET2, found among those with African ancestry, and the intergenic region between KPNA4 and TRIM59 which were associated with a 2.4 and a 1.16 fold increased risk of CHIP.(28) Further, a case control family study of patients with germline ATG2B/GSKIP-containing 14q23 duplication found that this was associated with a predisposition to develop TET2-related CHIP.(35) Additionally, a common inherited polymorphism in the TCL1A promoter was associated with slower clonal expansion. People carrying two protective alleles had up to 80% reduced odds of having driver mutations in TET2, ASXL1, SF3B1, SRSF2, and JAK2, but not DNMT3A.(36) More research is needed to better understand the possible inherited factors predisposing to CHIP.
In terms of modifiable risk factors, smoking has the most evidence of an association with CHIP (20,23,27,28,37–39), with one study showing that ASXL1-related CHIP has the greatest association with smoking (16,37). There are also some data suggesting an association between unhealthy diet (40) and vitamin C deficiency (41) predisposing to a higher prevalence of CHIP. Conversely, being normal weight, or overweight carried a lower odds (OR, 0.71 (95% CI, 0.57–0.88) and 0.83 (95% CI, 0.68–1.01), respectively) of developing CHIP compared to those who are obese (39). Inflammation may be another modifiable risk factor. For example, these hematopoietic stem cells (HSCs) may better resist inflammatory signals, leading to clonal selection in a pro-inflammatory environment.(42)
Some cancer treatments have also been linked to the development of CHIP. Cytotoxic chemotherapy has repeatedly been shown to be associated with an increased risk of CHIP. (16,20,24) Specifically, platinum chemotherapy, in particular carboplatin, (16,25,43) and topoisomerase II inhibitors (16) have been implicated in chemotherapy-associated CHIP, and result almost exclusively in TP53, PPM1D, and CHEK2 mutations (16,20,25). There was a dose response relationship between chemotherapy and CHIP suggesting that certain treatments are causally linked to CHIP.(16) Poly-ADP ribose polymerase (PARP) inhibitor therapy was also associated with an expansion of TP53-related CHIP clones among women with ovarian cancer.(43) Targeted therapies and immunotherapy with checkpoint inhibitors have not been found to be associated with the development or progression of CHIP.(16,22) Prior radiation, even short courses of radiation, have been found to increase the likelihood of developing CHIP (18,20).
Prevalence of CHIP among patients with solid tumor malignancies
CHIP is relatively common in patients with solid tumor malignancies. Four large studies have provided estimates from as low as 14% to as high as 65% prevalence among patients with solid tumors although most of these studies measure CHIP after having received treatment, which as illustrated above, could influence the presence of CHIP.(17–20) There is some evidence that certain cancers are more likely than others to be associated with CHIP. In a treatment naïve setting, patients with non-small cell lung cancer, breast cancer, pancreas, and prostate cancer have the highest prevalence of CHIP, while patients with renal cell carcinoma and ovarian cancer seem to have the lowest prevalence (16,44). Figure 2 summarizes the prevalence of CHIP across tumor types that have been estimated across studies, noting that the method for detecting CHIP varied by study. Whether or not the presence of CHIP is associated with an increased risk of the development of solid tumor malignancies is currently unknown. Certainly, given its association with the dysregulation of systemic inflammation, and the known association of inflammation with cancer, it is conceivable that it might be; however, this is currently an active area of research. (45,46) Furthermore, whether or not the mechanism of CHIP development in patients with different types of tumors is the same, and the same as the population without a cancer diagnosis, is also unknown.
Figure 2.
Prevalence of clonal hematopoiesis cited in the literature across different tumor types.
Clinical Implications for cancer risk and overall survival
CHIP has been associated with hematologic malignancies as well as non-malignant diseases. In the general population, CHIP is associated with an approximate 0.5% to 1% progression per year to an overt hematologic malignancy. (6,7,12) The malignant potential of CHIP is determined not only by the size of the clone but also by the type and number of mutated genes. The presence of mutations in spliceosome genes, IDH1/2 mutations, TP53 mutations, multiple mutations, and high variant allele frequencies are associated with an increased risk of myeloid malignancies. Interestingly, the risk appears lowest in patients with single DNMT3A or TET2 mutations. (47,48) While secondary hematologic malignancies are very rare among patients with solid tumor malignancies, the risk seems to be increased among those with CHIP. In one study, hematologic malignancies were more common among patients with CHIP (1% prevalence; 95% CI 0.5% - 1.8%) compared to solid tumor patients without a history of CHIP ( 0.3% prevalence; 95% 0.1–0.5%).(20) Case control studies found that between 62 to 71% of patients with solid tumor malignancies who did develop hematologic malignancies had antecedent CHIP compared to 27%–31% of control patients with solid tumor malignancies who did not develop hematologic malignancies.(49,50) How can we prospectively use this information? Breast cancer is one of the most common preceding malignancies to therapy-related myeloid neoplasms, an uncommon but incurable cancer with a very poor prognosis and median survival of 15 months.(51) Among women with a history of breast cancer who go on to develop therapy-related neoplasms, CHIP has been identified in tumor infiltrating leucocytes of the primary breast tumor.(52) This suggests that known pre-existing clonal hematopoiesis, especially in one of the high-risk therapy-related myeloid neoplasm genes, like TP53, may identify women at higher risk of developing hematologic malignancy following curative treatment for localized breast cancer. One study did assess if screening for CHIP can be used as a marker for the development of therapy-related myeloid neoplasms after breast cancer treatment (53). While this study did not show that CHIP was a risk factor, the median follow up time was 3.1 years, which is likely insufficient follow up to detect a difference given the median time from breast cancer treatment to therapy related myeloid neoplasm is 5–6 years. (52)
Consistently, clonal hematopoiesis is associated with lower overall survival in the general population, however, the relationship among patients with solid tumor malignancies is less clear.(6) When looking across cancers, CHIP has been associated with lower survival rates that were not driven by the development of hematologic malignancies.(20) There is a single study of metastatic colorectal cancer patients where CHIP was found to be an independent predictor of improved overall survival when considering age, treatment, extent of disease, and functional status.(24) This was most pronounced for patients with DNMT3A-mutated CHIP who also were found to have early tumor shrinkage, suggesting that there may be some relationship or interaction with certain treatments in certain cancers. (24) More research is needed to better understand if CHIP is a prognostic biomarker in patients with solid tumor malignancies, and if or how the presence of CHIP should be considered in the context of cancer clinical trials.
Clinical Implications for Nonmalignant Conditions
Outside of cancer, CHIP is associated with a number of conditions which have inflammation as a contributing etiology to the disease. The presence of CHIP is associated with 1.9 times the risk of atherosclerotic cardiovascular disease (95% CI 1.4, 2.7) and 4 times the risk of acute myocardial infarction (95% CI 2.4, 6.7) after adjusting for known cardiovascular disease risk factors.(54) This potentially has important implications for patients with solid tumor malignancies, as cardiovascular disease is a major cause of morbidity and mortality for patients with cancer.(55,56) Some therapies used for cancer treatment, for example androgen-targeting agents in prostate cancer, also increase the risk of cardiovascular disease.(57) As providers assess the risk-benefit ratio and choose certain treatments based on the side effect profile of the medications, it may be that knowing the presence of CHIP may help create a more personalized risk assessment for patients.
In addition to ischemic heart disease, CHIP is associated with stroke. In a study of 86,178 participants from 8 large cohort studies, CHIP was associated with a 14% increased risk of stroke after adjusting for age, race, and sex (HR 1.14, 95% CI 1.03 – 1.27), with a greater association with hemorrhagic stroke (HR 1.24 95% CI 1.01, 1.51) rather than ischemic stroke (HR 1.11, 0.98–1.25). This seems to vary by mutation. TET2 alterations were associated with a 90% increased risk of ischemic stroke (HR 1.18, 3.05) but no statistically significant increased risk of hemorrhagic stroke (HR 1.50, 0.87–2.61), whereas DNMT3A alterations were associated with an increased risk of hemorrhagic stroke (HR 1.44; 1.03–1.98) but no statistically significant increased risk of ischemic stroke (1.02; 0.82–1.26).(58) DMNT3A and TET2 mutations were both associated with an increased risk of hemorrhagic stroke. In the general population, CHIP has also been associated with a higher risk of heart failure,(59,60) COPD,(38,61) pulmonary hypertension,(62) autoimmune disease,(63) osteoporosis, and decreased bone mineral density.(64) Among oncology patients, CHIP was associated with an increased risk of infection – a nearly 2 times greater risk of severe COVID-19 disease, 2 times the risk of Clostridium difficile, and 56% increased risk of Streptococcus or Enterococcus infections.(65) Given the underlying associations with increased inflammation, it was hypothesized that CHIP would also contribute to the risk of VTE in oncology patients, however this has not been observed.(66)
Diagnostic Interference and misattribution
Much of the research presented above is based on the detection of CHIP for research purposes, identified by sequencing of peripheral blood mononuclear cells. In real world clinical practice, CHIP is most often inferred after being detected as an incidental finding on routine genetic testing. CHIP mutations identified on these tests can be misattributed as tumor or germline derived when in fact the mutations are somatic alterations in leukocytes.
Interference on germline genetic testing
Germline genetic testing in oncology patients is most commonly done via blood or saliva testing.(67) Results from blood samples can be contaminated by the presence of CHIP. Saliva-based samples may still contain a high proportion of white blood cells (rather than purely being buccal epithelial cells) although it varies considerably and depends on the sample acquisition technique.(68) The misattribution of germline alterations is rare, as germline alterations in many of the CHIP associated genes are associated with neurodevelopmental disorders. For example, ASXL1 germline variants are associated with Bohring-Opitz syndrome – a syndrome that presents with severe intellectual disability, developmental delay, and seizures.(69) There are some false positives where CHIP alterations are called germline genetic alterations. Among 6,060 patients undergoing germline genetic testing, pathogenic mutations were found to actually be CHIP in 18 of the patients, putting the prevalence of interference of all the patients at 0.3%, although CHIP was the most common incidental finding in this population.(70) Another study of 116,084 germline genetic tests found a prevalence of somatic interference in 0.05% of samples.(71) TP53 mutations were the most common in this cohort and besides TP53, ATM, and CHEK2, most of the genes associated with CHIP are not included on germline genetic testing panels.(71) However there are ways to distinguish between true germline alterations and CHIP interference. Among 84 patients who had germline blood or saliva-based testing where a pathogenic TP53 mutation was found, on further analysis, 11 of the 84 (13%) actually had CHIP and not true germline mutations in the TP53 gene. This was identified because patients did not meet the NCCN criteria for Li-Fraumani syndrome,(67) did not have a founder gene mutation that was identified, and subsequent testing was done, including obtaining the VAF which was much lower than would be expected for a germline alteration.(72) Therefore, among patients undergoing germline genetic testing, if testing identifies a pathogenic mutation in a cancer-associated gene but the family history does not fit with the clinical picture, or the variant allele frequency is low (i.e. < 25%), interference from CHIP could be the likely explanation.
Interference on somatic testing
Interference of CHIP in the interpretation of somatic samples is more common. The rate of blood contamination in biopsy and surgical specimens contributes to the likelihood of this, and as the tumor purity of the sample decreases, the prevalence of CHIP increases.(73) The detection of CHIP in tumor specimens is associated with an increase in lymphocytes in the tumor microenvironment but it is unclear if this is because of the association of CHIP with inflammation or if this is just due to more admixing of the sample under study.(73) This occurs in an estimate of 5% to 29% of tissue specimens sent for next-generation sequencing.(21,22,74) Among patients who are known to have CHIP, 77% of them had the CHIP variant detected in the tumor tissue.(20) Variant allele frequencies may be helpful as they should vary between that found in blood (median in one study of 4.4%) compared to the VAF in the corresponding individual’s tumor tissue (median 0.5%).(20)
Perhaps the most problematic interference occurs in the setting of liquid biopsies. Liquid biopsies are increasingly being studied and used in clinical practice. Estimates are that around 50% of mutations identified in cell free DNA actually arise from CHIP rather than tumor.(13,75) Depending on the gene alteration, this may or may not have clinical implications. In one study of cell free DNA from 69 patients with prostate cancer, 10% of the patients were found to have CHIP mutations in homologous recombination repair genes. In this study, the simultaneous use of whole-blood controls largely mitigated the problem of mistaking CHIP as tumor-derived mutations.(76) Another study of 58 patients with lung cancer however demonstrated that KRAS mutations due to CHIP may also occur but are less common (3%).(77) This is important because these results would have been considered to be tumor-derived and therefore make the patient eligible to receive certain treatments; in the first example, it would have made patients eligible to receive PARP inhibitor treatment.(76) However, there would be no anticipated benefit to the patient when taking a drug to target a somatic, presumed tumor derived, mutation when in fact the gene of interest is actually CHIP-derived. Alternatively, the identification of CHIP could also lead to misinterpretation of a cancer recurrence and potentially lead to more aggressive, but unnecessary, treatment. These scenarios highlight the ways that CHIP interference on liquid biopsies can be misleading to clinicians.
Looking ahead
There are no formal recommendations for what to do when true clonal hematopoiesis is identified for patients with solid tumor malignancies. In the setting of otherwise unexplained cytopenias, some have suggested a bone marrow biopsy to evaluate for an underlying hematologic neoplasm.(78) Regular monitoring of complete blood counts to look for evidence of clinically significant cytopenias or progression to malignant neoplasm have also been suggested.(78,79) The management of cardiovascular risk factors may also be changed based on the presence of clonal hematopoiesis.(80) Specialized multi-disciplinary clinics are also being developed including participation by cardiologists, geneticists, oncologists, counselors, social workers, and other specialized services.(81) More research is needed about the differential risk of cancer and non-cancer diseases based on the type of CHIP mutation identified. Additional research is also needed on what, if any, clinical implications there are for cancer treatment when CHIP is found. For example, in patients with TP53-associated CHIP which is associated with hematologic malignancies, the type of chemotherapy or radiation used in the treatment of a primary malignancy might be changed to an option with a lower risk of therapy-related myeloid neoplasms. Alternatively, in a man with advanced prostate cancer who is deciding on whether or not to start androgen axis targeting agents, the presence of TET2-mutated CHIP might influence treatment decisions because treatment and CHIP can be associated with cardiovascular disease. Whether or not the presence of CHIP influences clinical outcomes from cancer treatments, including from immunotherapy, is an active area of research.
The presence of CHIP can have broad clinical consequences, from interference on next-generation sequencing results, to clinical consequences. In liquid biopsies, it will be increasingly important to distinguish CHIP mutations from mutations of tumor origin by improving and standardizing the technical processes. Future research is needed to determine how solid tumor malignancies and their treatments alter the progression of CHIP, and in turn, how CHIP might be used to improve treatment selection and patient outcomes.
Summarize key findings.
Clonal hematopoiesis of indeterminate potential (CHIP) is common among individuals with solid tumor malignancies.
CHIP is associated with an increased risk of developing hematologic malignancies and non-hematologic diseases like cardiovascular disease, and other inflammatory conditions
CHIP can interfere with the interpretation of next generation DNA sequencing results in patients with solid tumors
Acknowledgments
This work is partially supported by the National Institutes of Health Cancer Center Support Grants P30 CA006973, V Foundation, and the Prostate Cancer Foundation.
REFERENCES
- 1.Doulatov S, Notta F, Laurenti E, Dick JE. Hematopoiesis: A Human Perspective. Cell Stem Cell. 2012;10:120–36. [DOI] [PubMed] [Google Scholar]
- 2.Lee-Six H, Øbro NF, Shepherd MS, Grossmann S, Dawson K, Belmonte M, et al. Population dynamics of normal human blood inferred from somatic mutations. Nature. 2018;561:473–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Welch JS, Ley TJ, Link DC, Miller CA, Larson DE, Koboldt DC, et al. The origin and evolution of mutations in acute myeloid leukemia. Cell. 2012;150:264–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Steensma DP, Bejar R, Jaiswal S, Lindsley RC, Sekeres MA, Hasserjian RP, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood. 2015;126:9–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Midic D, Rinke J, Perner F, Müller V, Hinze A, Pester F, et al. Prevalence and dynamics of clonal hematopoiesis caused by leukemia-associated mutations in elderly individuals without hematologic disorders. Leukemia. 2020;34:2198–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, et al. Age-Related Clonal Hematopoiesis Associated with Adverse Outcomes. N Eng J Med. 2014;371:2488–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Genovese G, Kähler AK, Handsaker RE, Lindberg J, Rose SA, Bakhoum SF, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014;371:2477–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Watson CJ, Papula AL, Poon GYP, Wong WH, Young AL, Druley TE, et al. The evolutionary dynamics and fitness landscape of clonal hematopoiesis. Science. 2020;367:1449–54. [DOI] [PubMed] [Google Scholar]
- 9.Young AL, Challen GA, Birmann BM, Druley TE. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat Commun. 2016;7:12484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Jaiswal S, Ebert BL. Clonal hematopoiesis in human aging and disease. Science. 2019;366:eaan4673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Challen GA, Goodell MA. Clonal hematopoiesis: mechanisms driving dominance of stem cell clones. Blood. 2020;136:1590–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zink F, Stacey SN, Norddahl GL, Frigge ML, Magnusson OT, Jonsdottir I, et al. Clonal hematopoiesis, with and without candidate driver mutations, is common in the elderly. Blood. 2017;130:742–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Razavi P, Li BT, Brown DN, Jung B, Hubbell E, Shen R, et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat Med. 2019;25:1928–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Poon GYP, Watson CJ, Fisher DS, Blundell JR. Synonymous mutations reveal genome-wide levels of positive selection in healthy tissues. Nat Genet. 2021;53:1597–605. [DOI] [PubMed] [Google Scholar]
- 15.Mitchell E, Spencer Chapman M, Williams N, Dawson KJ, Mende N, Calderbank EF, et al. Clonal dynamics of haematopoiesis across the human lifespan. Nature. 2022;606:343–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bolton KL, Ptashkin RN, Gao T, Braunstein L, Devlin SM, Kelly D, et al. Cancer therapy shapes the fitness landscape of clonal hematopoiesis. Nat Genet. 2020;52:1219–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Zhang Y, Yao Y, Xu Y, Li L, Gong Y, Zhang K, et al. Pan-cancer circulating tumor DNA detection in over 10,000 Chinese patients. Nat Commun. 2021;12:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Coombs CC, Gillis NK, Tan X, Berg JS, Ball M, Balasis ME, et al. Identification of Clonal Hematopoiesis Mutations in Solid Tumor Patients Undergoing Unpaired Next-Generation Sequencing Assays. Clin Cancer Res. 2018;24:5918–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Li Z, Huang W, Yin JC, Na C, Wu X, Shao Y, et al. Comprehensive next-generation profiling of clonal hematopoiesis in cancer patients using paired tumor-blood sequencing for guiding personalized therapies. Clin Transl Med. 2020;10:e222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Coombs CC, Zehir A, Devlin SM, Kishtagari A, Syed A, Jonsson P, et al. Therapy-Related Clonal Hematopoiesis in Patients with Non-hematologic Cancers Is Common and Associated with Adverse Clinical Outcomes. Cell Stem Cell. 2017;21:374–382.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ptashkin RN, Mandelker DL, Coombs CC, Bolton K, Yelskaya Z, Hyman DM, et al. Prevalence of Clonal Hematopoiesis Mutations in Tumor-Only Clinical Genomic Profiling of Solid Tumors. JAMA Oncol. 2018;4:1589–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Miller PG, Gibson CJ, Mehta A, Sperling AS, Frederick DT, Manos MP, et al. Fitness Landscape of Clonal Hematopoiesis Under Selective Pressure of Immune Checkpoint Blockade. JCO Precis Oncol. 2020;4:1027–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hong W, Li A, Liu Y, Xiao X, Christiani DC, Hung RJ, et al. Clonal Hematopoiesis Mutations in Lung Cancer Patients are Associated with Lung Cancer Risk Factors. Cancer Res. 2021;canres.1903.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Arends CM, Dimitriou S, Stahler A, Hablesreiter R, Strzelecka PM, Stein CM, et al. Clonal hematopoiesis associates with improved survival in metastatic colorectal cancer patients from the FIRE-3 trial. Blood. 2021; [DOI] [PubMed] [Google Scholar]
- 25.Weber-Lassalle K, Ernst C, Reuss A, Möllenhoff K, Baumann K, Jackisch C, et al. Clonal Hematopoiesis-Associated Gene Mutations in a Clinical Cohort of 448 Patients with Ovarian Cancer. J Natl Cancer Inst. 2021;djab231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Xie M, Lu C, Wang J, McLellan MD, Johnson KJ, Wendl MC, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med. 2014;20:1472–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Saiki R, Momozawa Y, Nannya Y, Nakagawa MM, Ochi Y, Yoshizato T, et al. Combined landscape of single-nucleotide variants and copy number alterations in clonal hematopoiesis. Nat Med. 2021;27:1239–49. [DOI] [PubMed] [Google Scholar]
- 28.Bick AG, Weinstock JS, Nandakumar SK, Fulco CP, Bao EL, Zekavat SM, et al. Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature. 2020;1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Rossi M, Meggendorfer M, Zampini M, Tettamanti M, Riva E, Travaglino E, et al. Clinical relevance of clonal hematopoiesis in persons aged ≥80 years. Blood. 2021;138:2093–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Nachun D, Lu AT, Bick AG, Natarajan P, Weinstock J, Szeto MD, et al. Clonal hematopoiesis associated with epigenetic aging and clinical outcomes. Aging Cell. 2021;20:e13366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Honigberg MC, Zekavat SM, Niroula A, Griffin GK, Bick AG, Pirruccello JP, et al. Premature Menopause, Clonal Hematopoiesis, and Coronary Artery Disease in Postmenopausal Women. Circulation. 2021;143:410–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hansen JW, Pedersen DA, Larsen LA, Husby S, Clemmensen SB, Hjelmborg J, et al. Clonal hematopoiesis in elderly twins: concordance, discordance, and mortality. Blood. 2020;135:261–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Fabre MA, McKerrell T, Zwiebel M, Vijayabaskar MS, Park N, Wells PM, et al. Concordance for clonal hematopoiesis is limited in elderly twins. Blood. 2020;135:269–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Nakao T, Bick AG, Taub MA, Zekavat SM, Uddin MM, Niroula A, et al. Mendelian randomization supports bidirectional causality between telomere length and clonal hematopoiesis of indeterminate potential. Sci Adv. 2022;8:eabl6579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Pegliasco J, Hirsch P, Marzac C, Isnard F, Meniane J-C, Deswarte C, et al. Germline ATG2B/GSKIP-containing 14q32 duplication predisposes to early clonal hematopoiesis leading to myeloid neoplasms. Leukemia. 2022;36:126–37. [DOI] [PubMed] [Google Scholar]
- 36.Weinstock JS, Gopakumar J, Burugula BB, Uddin MM, Jahn N, Belk JA, et al. Clonal hematopoiesis is driven by aberrant activation of TCL1A. bioRxiv. 2021;2021.12.10.471810. [Google Scholar]
- 37.Dawoud AAZ, Tapper WJ, Cross NCP. Clonal myelopoiesis in the UK Biobank cohort: ASXL1 mutations are strongly associated with smoking. Leukemia. 2020;34:2660–72. [DOI] [PubMed] [Google Scholar]
- 38.Miller PG, Qiao D, Rojas-Quintero J, Honigberg MC, Sperling AS, Gibson CJ, et al. Association of clonal hematopoiesis with chronic obstructive pulmonary disease. Blood. 2022;139:357–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Haring B, Reiner AP, Liu J, Tobias DK, Whitsel E, Berger JS, et al. Healthy Lifestyle and Clonal Hematopoiesis of Indeterminate Potential: Results From the Women’s Health Initiative. J Am Heart Assoc. 2021;10:e018789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bhattacharya R, Zekavat SM, Uddin MM, Pirruccello J, Niroula A, Gibson C, et al. Association of Diet Quality With Prevalence of Clonal Hematopoiesis and Adverse Cardiovascular Events. JAMA Cardiol. 2021;6:1069–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chen J, Nie D, Wang X, Wang L, Wang F, Zhang Y, et al. Enriched clonal hematopoiesis in seniors with dietary vitamin C inadequacy. Clin Nutr ESPEN. 2021;46:179–84. [DOI] [PubMed] [Google Scholar]
- 42.Avagyan S, Henninger JE, Mannherz WP, Mistry M, Yoon J, Yang S, et al. Resistance to inflammation underlies enhanced fitness in clonal hematopoiesis. Science. 2021;374:768–72. [DOI] [PubMed] [Google Scholar]
- 43.Kwan TT, Oza AM, Tinker AV, Ray-Coquard I, Oaknin A, Aghajanian C, et al. Preexisting TP53-Variant Clonal Hematopoiesis and Risk of Secondary Myeloid Neoplasms in Patients With High-grade Ovarian Cancer Treated With Rucaparib. JAMA Oncology. 2021;7:1772–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Marshall CH, Holler AE, Tsai H-L, Gondek L, Luo J, Antonarakis ES. Clonal hematopoiesis in prostate cancer inferred from somatic tumor profiling. JCO. 2021;39:e17001–e17001. [Google Scholar]
- 45.Jaiswal S, Libby P. Clonal haematopoiesis: connecting ageing and inflammation in cardiovascular disease. Nat Rev Cardiol. 2020;17:137–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. [DOI] [PubMed] [Google Scholar]
- 47.Abelson S, Collord G, Ng SWK, Weissbrod O, Mendelson Cohen N, Niemeyer E, et al. Prediction of acute myeloid leukaemia risk in healthy individuals. Nature. 2018;559:400–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Desai P, Mencia-Trinchant N, Savenkov O, Simon MS, Cheang G, Lee S, et al. Somatic mutations precede acute myeloid leukemia years before diagnosis. Nat Med. 2018;24:1015–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Takahashi K, Wang F, Kantarjian H, Doss D, Khanna K, Thompson E, et al. Preleukaemic clonal haemopoiesis and risk of therapy-related myeloid neoplasms: a case-control study. Lancet Oncol. 2017;18:100–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Gillis NK, Ball M, Zhang Q, Ma Z, Zhao Y, Yoder SJ, et al. Clonal haemopoiesis and therapy-related myeloid malignancies in elderly patients: a proof-of-concept, case-control study. Lancet Oncol. 2017;18:112–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Fianchi L, Pagano L, Piciocchi A, Candoni A, Gaidano G, Breccia M, et al. Characteristics and outcome of therapy-related myeloid neoplasms: Report from the Italian network on secondary leukemias. Amer J Hematology. 2015;90:E80–5. [DOI] [PubMed] [Google Scholar]
- 52.Comen EA, Bowman RL, Selenica P, Kleppe M, Farnoud NR, Pareja F, et al. Evaluating Clonal Hematopoiesis in Tumor-Infiltrating Leukocytes in Breast Cancer and Secondary Hematologic Malignancies. JNCI. 2020;112:107–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Slovak ML, Bedell V, Lew D, Albain KS, Ellis GK, Livingston RB, et al. Screening for clonal hematopoiesis as a predictive marker for development of therapy-related myeloid neoplasia (t-MN) following neoadjuvant therapy for breast cancer: a Southwest Oncology Group study (S0012). Breast Cancer Res Treat. 2010;119:391–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N Eng J Med. 2017;377:111–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zaorsky NG, Churilla TM, Egleston BL, Fisher SG, Ridge JA, Horwitz EM, et al. Causes of death among cancer patients. Ann Oncol. 2017;28:400–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Stoltzfus KC, Zhang Y, Sturgeon K, Sinoway LI, Trifiletti DM, Chinchilli VM, et al. Fatal heart disease among cancer patients. Nat Commun. 2020;11:2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Hu J-R, Duncan MS, Morgans AK, Brown JD, Meijers WC, Freiberg MS, et al. Cardiovascular Effects of Androgen Deprivation Therapy in Prostate Cancer: Contemporary Meta-Analyses. Arterioscler Thromb Vasc Biol. 2020;40:e55–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bhattacharya R, Zekavat SM, Haessler J, Fornage M, Raffield L, Uddin MM, et al. Clonal Hematopoiesis Is Associated With Higher Risk of Stroke. Stroke. American Heart Association; 0:STROKEAHA.121.037388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Yu B, Roberts MB, Raffield LM, Zekavat SM, Nguyen NQH, Biggs ML, et al. Supplemental Association of Clonal Hematopoiesis With Incident Heart Failure. J Am Coll Cardiol. 2021;78:42–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Pascual-Figal DA, Bayes-Genis A, Díez-Díez M, Hernández-Vicente Á, Vázquez-Andrés D, de la Barrera J, et al. Clonal Hematopoiesis and Risk of Progression of Heart Failure With Reduced Left Ventricular Ejection Fraction. J Am Coll Cardiol. 2021;77:1747–59. [DOI] [PubMed] [Google Scholar]
- 61.van Zeventer IA, Salzbrunn JB, de Graaf AO, van der Reijden BA, Boezen HM, Vonk JM, et al. Prevalence, predictors, and outcomes of clonal hematopoiesis in individuals aged ≥80 years. Blood Adv. 2021;5:2115–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kimishima Y, Misaka T, Yokokawa T, Wada K, Ueda K, Sugimoto K, et al. Clonal hematopoiesis with JAK2V617F promotes pulmonary hypertension with ALK1 upregulation in lung neutrophils. Nat Commun. 2021;12:6177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hecker JS, Hartmann L, Rivière J, Buck MC, van der Garde M, Rothenberg-Thurley M, et al. CHIP and hips: clonal hematopoiesis is common in patients undergoing hip arthroplasty and is associated with autoimmune disease. Blood. 2021;138:1727–32. [DOI] [PubMed] [Google Scholar]
- 64.Kim PG, Niroula A, Shkolnik V, McConkey M, Lin AE, Słabicki M, et al. Dnmt3a-mutated clonal hematopoiesis promotes osteoporosis. J Exp Med. 2021;218:e20211872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Bolton KL, Koh Y, Foote MB, Im H, Jee J, Sun CH, et al. Clonal hematopoiesis is associated with risk of severe Covid-19. Nat Commun.2021;12:5975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Dunbar A, Bolton KL, Devlin SM, Sanchez-Vega F, Gao J, Mones JV, et al. Genomic profiling identifies somatic mutations predicting thromboembolic risk in patients with solid tumors. Blood. 2021;137:2103–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Daly MB, Pal T, Berry MP, Buys SS, Dickson P, Domchek SM, et al. Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network. National Comprehensive Cancer Network; 2021;19:77–102. [DOI] [PubMed] [Google Scholar]
- 68.Chao EC, Astbury C, Deignan JL, Pronold M, Reddi HV, Weitzel JN, et al. Incidental detection of acquired variants in germline genetic and genomic testing: a points to consider statement of the American College of Medical Genetics and Genomics (ACMG). Genet Med. 2021;23:1179–84. [DOI] [PubMed] [Google Scholar]
- 69.Brunet T, Berutti R, Dill V, Hecker JS, Choukair D, Andres S, et al. Clonal Hematopoiesis as a pitfall in germline variant interpretation in the context of Mendelian disorders. Hum Mol Genet. 2022;ddac034. [DOI] [PubMed] [Google Scholar]
- 70.Maani N, Panabaker K, McCuaig JM, Buckley K, Semotiuk K, Farncombe KM, et al. Incidental findings from cancer next generation sequencing panels. Genom Med. 2021;6:1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Weitzel JN, Chao EC, Nehoray B, Van Tongeren LR, LaDuca H, Blazer KR, et al. Somatic TP53 variants frequently confound germ-line testing results. Genet Med. 2018;20:809–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Schwartz AN, Hyman SR, Stokes SM, Castillo D, Tung NM, Weitzel JN, et al. Evaluation of TP53 Variants Detected on Peripheral Blood or Saliva Testing: Discerning Germline From Somatic TP53 Variants. JCO Precis Oncol. 2021;5:1677–86. [DOI] [PubMed] [Google Scholar]
- 73.Severson EA, Riedlinger GM, Connelly CF, Vergilio J-A, Goldfinger M, Ramkissoon S, et al. Detection of clonal hematopoiesis of indeterminate potential in clinical sequencing of solid tumor specimens. Blood. 2018;131:2501–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Chan HT, Nagayama S, Chin YM, Otaki M, Hayashi R, Kiyotani K, et al. Clinical significance of clonal hematopoiesis in the interpretation of blood liquid biopsy. Mol Oncol. 2020;14:1719–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Antonarakis ES, Kuang Z, Tukachinsky H, Parachoniak C, Kelly AD, Gjoerup O, et al. Prevalence of inferred clonal hematopoiesis (CH) detected on comprehensive genomic profiling (CGP) of solid tumor tissue or circulating tumor DNA (ctDNA). JCO. 2021;39:3009–3009. [Google Scholar]
- 76.Jensen K, Konnick E, Schweizer MT, Sokolova AO, Grivas P, Cheng HH, et al. Association of Clonal Hematopoiesis in DNA Repair Genes With Prostate Cancer Plasma Cell-free DNA Testing Interference. JAMA Oncology. 2020;7:107–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Hu Y, Ulrich BC, Supplee J, Kuang Y, Lizotte PH, Feeney NB, et al. False-Positive Plasma Genotyping Due to Clonal Hematopoiesis. Clin Cancer Res. 2018;24:4437–43. [DOI] [PubMed] [Google Scholar]
- 78.Bolton KL, Gillis NK, Coombs CC, Takahashi K, Zehir A, Bejar R, et al. Managing Clonal Hematopoiesis in Patients With Solid Tumors. J Clin Oncol. 2019;37:7–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Gondek LP, DeZern AE. Assessing clonal haematopoiesis: clinical burdens and benefits of diagnosing myelodysplastic syndrome precursor states. Lancet Haematol. 2020;7:e73–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sidlow R, Lin AE, Gupta D, Bolton KL, Steensma DP, Levine RL, et al. The Clinical Challenge of Clonal Hematopoiesis, a Newly Recognized Cardiovascular Risk Factor. JAMA Cardiology. 2020;5:958–61. [DOI] [PubMed] [Google Scholar]
- 81.Steensma DP, Bolton KL. What to tell your patient with clonal hematopoiesis and why: insights from 2 specialized clinics. Blood. 2020;136:1623–31. [DOI] [PMC free article] [PubMed] [Google Scholar]