Clonal hematopoiesis

Last updated

Clonal hematopoiesis of indeterminate potential, or CHIP, is a common aging-related phenomenon in which hematopoietic stem cells (HSCs) or other early blood cell progenitors contribute to the formation of a genetically distinct subpopulation of blood cells. [1] [2] [3] As the name suggests, this subpopulation in the blood is characterized by a shared unique mutation in the cells' DNA; it is thought that this subpopulation is "clonally" derived from a single founding cell and is therefore made of genetic "clones" of the founder. [4] [5] [6] [7] The establishment of a clonal population may occur when a stem or progenitor cell acquires one or more somatic mutations that give it a competitive advantage in hematopoiesis over the stem/progenitor cells without these mutations. [1] [3] Alternatively, clonal hematopoiesis may arise without a driving mutation, through mechanisms such as neutral drift in the stem cell population. [8] Clonal hematopoiesis may occur in people who are completely healthy but has also been found in people with hematologic diseases. [1] [9] [10] The clonal population may vary in size depending on the person, where it can be less than 2% of the blood or, at the other end, can sometimes grow close to 100%. [4] [9] The incidence of clonal hematopoiesis has been found to rise dramatically with age. Recent studies have demonstrated that less than 1% of the population under age 40 but approximately 10-20% of the population over age 70 has observable clonal hematopoiesis. [4] [5] [6] Having clonal hematopoiesis has been linked to a more than 10-fold increased risk of developing a blood cancer, though the overall likelihood is still low. [4] [5] Clonal hematopoiesis does not typically give rise to noticeable symptoms, but does lead to increased risk of cardiovascular disease. [1] [5] [11] Patients with solid tumors or lymphoma and clonal hematopoiesis have been shown to have an inferior outcome. [12]

Contents

History

The first major evidence for the existence of prevalent clonal hematopoiesis in healthy people was put forth in the 1990s. Using the HUMARA assay, scientists found that there was nonrandom X-inactivation of the X chromosome in the blood of some healthy women. [13] [14] This means that a greater than expected proportion of the blood had the silencing of one specific X chromosome in the chromosome pair. Just as the observation of the same DNA mutation in a subset of cells suggests a single founding source, this X-inactivation skew suggests that a greater than expected number of cells are being generated from the same precursor. Importantly, these findings described an increase in this nonrandom skewing with increasing age, hinting that unobserved mutations acquired with age could be driving a clonal expansion. In a similar vein, other studies using the HUMARA technology had found that hematologic malignancies are clonal diseases even when there is no apparent chromosomal abnormality, [15] [16] and that there are pre-leukemic clonal populations which precede acute myeloid leukemia (AML). [17] As the HUMARA assay is based on the epigenetic state of cells, the underlying genetic determinants of the clonal expansion remained to be uncovered.

This set of evidence led to the suggestion in 2005 that driving mutations in leukemia are acquired in a step-wise manner. [18] This model has received support from studies showing subpopulations of blood cells harboring initiating but not late somatic mutations in patients with chronic lymphocytic leukemia (CLL), [19] [20] hairy cell leukemia (HCL), [21] and AML. [22] [23] [24]

The combination of these two ideas, that clonal hematopoiesis might be common in the elderly population and that AML evolves from pre-leukemic populations, led to the hypothesis that malignancy-associated mutations could also contribute to asymptomatic clonal hematopoiesis in healthy individuals. [1] This view gained mechanistic support in 2012 when it was found a number of the women who showed evidence for clonal hematopoiesis through X-inactivation skew also had mutations in the hematologic-malignancy-associated gene TET2. [25]

In 2014, several independent studies confirmed the presence of malignancy-associated mutations in the blood of individuals who have no clinical signs of hematologic malignancy. [4] [5] [6] In combination, these studies have demonstrated the widespread incidence of clonal hematopoiesis in the healthy adult population and have stimulated further efforts to broaden our understanding of clonal hematopoiesis in health and disease. The term "clonal hematopoiesis of indeterminate potential" (CHIP) was proposed later that year to describe persons who do not have a malignancy meeting World Health Organization diagnostic criteria, yet have somatic mutations in hematopoietic stem and progenitor cells involving genes that have been associated with hematological malignancy, and these mutations are present in blood cells with a variant allele frequency of at least 2%. [3] The 2% threshold was chosen in part because of technical limitations (i.e., analytic sensitivity of clinically available sequencing assays) but also because very small clones are of unclear clinical significance.

Population genetics

The advent of next-generation DNA sequencing has allowed for the targeted identification of somatic mutations involved in clonal hematopoiesis at the population level. The studies undertaken as of 2017 are largely consistent in their main findings. One common finding has been that observable clonal hematopoiesis is virtually absent from the under-40 population, with a sharp uptick in frequency past 60 years of age. [4] [5] [6] Indeed, the evidence from these studies suggests that between 10% and 20% of the population over age 70 have clonal hematopoiesis. In the U.S. alone, this means that, at the low end, some 2,975,000 seniors over 70 years of age are living with this condition. [26]

Number of people estimated to be affected by clonal hematopoiesis out of the total U.S. population, by age group. Clonal Hematopoiesis in US 2015 Estimate.jpg
Number of people estimated to be affected by clonal hematopoiesis out of the total U.S. population, by age group.

The other main common finding is that there are many different mutations involved in clonal hematopoiesis. Many of these fall into the categories of epigenetic regulators (DNMT3a, TET2, and ASXL1 ), signaling proteins (JAK2), spliceosome components ( SF3B1 and SRSF2), or members of the DNA damage response ( TP53 and PPM1D ). [4] [5] [6] Many people identified as having clonal hematopoiesis have a mutation in a single gene, though a significant number have mutations in two or more genes. [4] [5] [6] The number and variety of observed mutations suggests that these mutations may contribute to clonal hematopoiesis by several distinct mechanisms, discussed in more detail below.

There is also limited evidence suggesting clonal hematopoiesis may be ubiquitous in healthy adults, albeit at extremely low levels (less than 0.1% of peripheral blood cells). A study employing the ultra-sensitive digital droplet PCR method found that 95% of studied individuals (19 out of 20) between the ages of 50 and 70 had at least low-level clonal hematopoiesis. [27] This finding does not necessarily conflict with earlier reports that clonal hematopoiesis is not ubiquitous in this age bracket, as these previous studies' experimental designs compels the use of a higher threshold to identify legitimate clonal hematopoiesis. [4] [5] [6]

Ongoing studies are examining what genetic and epidemiological factors may influence the acquisition of mutations in clonal hematopoiesis.

Biology

Clonal hematopoiesis is thought to originate with the hematopoietic stem cells that make blood. An adult human has approximately 10,000 to 20,000 HSCs. [28] The fact that these cells are maintained for life and each HSC may acquire about one mutation in a protein-coding exon each decade [29] means that an elderly individual will have a certain amount of genetic mosaicism, or a variety of cells with different unique mutations, within their HSC population. However, this does not lead to clonal hematopoiesis in all cases. It is only when the genetic mutation confers a selective advantage on its host or there is another favorable stem cell dynamic that there is a clonal expansion.

Candidate driver mutations

There are several general mechanisms by which a mutation could provide such an advantage and it is likely that the mutations found in clonal hematopoiesis act through different pathways. First, a mutation could provide a growth advantage, causing HSCs to divide more rapidly and contribute a larger proportion of the mature blood cells. This may be the case for mutations in genes related to signaling, such as that which causes the activating V617F substitution in the JAK2 signaling protein. Mutations in the DNA damage response genes would appear more likely to act via a second mechanism: allowing for HSC survival and proliferation under normally lethal cytotoxic stress. [1]

Other mechanisms are more likely to be associated with the disruption of epigenetic regulators, which comprises 80% of observed mutations in clonal hematopoiesis. A third potential mechanism of action is that the mutation makes the HSC-derived progenitor cells less able to differentiate into mature blood cells. This would allow these cells to continue to divide even after they would have normally stopped, since progenitor cells may divide whereas normal mature blood cells cannot. A fourth possibility is that the mutation makes the progenitor cells and cells derived from them more like stem cells in their ability to keep dividing. The previous two possibilities are very similar in terms of physiologic outcome and mainly differ on what is happening at the DNA level: whether differentiation genes are suppressed or a stem cell program is upregulated. A final possibility is that a gradient of epigenetic states is created in the HSC and progenitor cells and the cells with the most favorable epigenetics are able to grow out faster than unmutated cells. [1]

Non-candidate-driver mechanisms

An expansion of blood cells from a single source does not necessarily require a mutation to act as the driving force. A large proportion of the population who exhibit clonal hematopoiesis have no identifiable mutations in known candidate driver genes. [4] [8] One possible explanation is that among a naturally-occurring spectrum of inheritable epigenetic states, there are those which augment the self-renewal or proliferation of a stem cell and its progeny. [8] Another explanation is that a process of neutral drift causes the predominance of a clonal stem cell population over time. In this scenario, all stem cells have an equal proliferative potential but some of them die out in a stochastic manner leading some of the remaining cells to proliferate to replace them. [8] [30] This can be equated to a game of chance where all players start with the same odds of winning. As the game is played, winners and losers will arise despite the equal starting positions. [30]

Implications for human health

Clonal hematopoiesis by itself is not considered to be a hematologic cancer; nevertheless, evidence is mounting that this condition may adversely affect human health. It has been proposed to label the group of individuals who have clonal hematopoiesis defined by a mutation in a malignancy-associated gene but without evidence of disease (such as cytopenia, dysplasia or immature "blast" cells in the bone marrow) as having Clonal Hematopoiesis of Indeterminate Potential (CHIP). [1] [3] [31] A clonal involvement (sometimes referred to simply as the size of a "clone") of 2% of the blood has been tentatively proposed as a cutoff, though there is discussion that a lower floor that is more inclusive could also be appropriate. [1] [3] [27] [32] This cutoff may ultimately depend on whether clones must reach a certain size before influencing health. The level at which a clone begins to have a potential clinical impact is an open question, though there is already data to suggest larger clones have a larger effect on health. [5]

The presence of clonal hematopoiesis/CHIP has been shown to increase blood cancer risk and is correlated with an increased risk of mortality overall. [4] [5] [8] This is true both of clonal hematopoiesis with known candidate drivers as well as in cases without such drivers. [8]

Blood cancer risk

One area of health that CHIP has been definitively shown to influence is the risk of progression to blood cancer. In a given year, a tiny fraction of the general population will develop a hematologic cancer such as myelodysplastic syndrome (MDS) or AML; it is estimated that just 3 to 4 people per 100,000 will get MDS in a given year, [33] and 4 people per 100,000 will develop AML. [34] With CHIP, the risk of acquiring a hematologic malignancy like MDS or AML is increased more than 10-fold. [4] [5] Despite this increased risk, people with CHIP are still at low overall risk for developing a blood cancer, with only about 0.5-1.0% transformation per year. [1]

Cardiovascular risk

A second area of health that may be affected by CHIP is the risk for heart attack and stroke. A strong association between CHIP and heart attack/ischemic stroke has been identified in one human genetic dataset, where CHIP was a stronger predictor of heart attack/stroke than if a patient was a smoker, had hypertension, had high cholesterol, or was overweight. In this study, which shows correlation but not causation, people with CHIP were 2.3 times more likely to have a heart attack, or 4.4 times as likely if the variant allele frequency in their blood was greater than 0.10, than matched controls without CHIP. [5] It has also been found that there is an increased risk of cardiovascular mortality in patients who exhibit CHIP and receive self-derived stem cell transplantation. [10] In addition to heart attack and stroke, human studies further suggest an association of CHIP with heart failure and cardiac arrhythmias. [35] [36]

The idea of CHIP having a causal role in human heart attacks/strokes has been given support by a 2017 study that showed impairment of the Tet2 CHIP gene in mice causally led to accelerated atherosclerosis, [37] and this finding in mice has been independently validated. [11] The possibility of somatic mutations in the blood contributing not only to cancer risk but also to heart attack and stroke has generated much discussion in top-level scientific publications [38] [39] and a large multi-cohort study published in 2017 appears to confirm the causal link between CHIP and cardiovascular disease in humans. [11]

Comorbidities

In addition to its effects on those who would otherwise be considered healthy, CHIP may have implications in certain disease contexts. It has been shown that patients with CHIP who receive autologous stem cell transplantation (ASCT) as part of their treatment for lymphoma have worse outcomes than patients without CHIP. The poorer prognosis for these patients is due to both an increase in subsequent therapy-related myeloid neoplasms and increased risk for cardiovascular mortality. [10]

Treatment

There are currently no therapies for slowing or targeting CHIP mutations. Together with the fact that progression from CHIP to outright hematologic malignancy remains infrequent, medical experts have argued against preemptive screening for CHIP but suggest routine follow-up for incidental CHIP findings. [1] [3]

Associated disorders

Clonal hematopoiesis is sometimes compared to the unrelated blood disorders of monoclonal gammopathy of undetermined significance (MGUS) and monoclonal B-cell lymphocytosis (MBL) to which it bears similarities in its apparent priming for more advanced hematologic disease combined with a lack of symptoms and overall low risk of progression. [1] [3] The acquisition of additional mutations can cause CHIP to transform into the related blood disorders MDS and AML. [3] [31]

See also

Related Research Articles

<span class="mw-page-title-main">Haematopoiesis</span> Formation of blood cellular components

Haematopoiesis is the formation of blood cellular components. All cellular blood components are derived from haematopoietic stem cells. In a healthy adult human, roughly ten billion to a hundred billion new blood cells are produced per day, in order to maintain steady state levels in the peripheral circulation.

<span class="mw-page-title-main">Myelodysplastic syndrome</span> Diverse collection of blood-related cancers

A myelodysplastic syndrome (MDS) is one of a group of cancers in which immature blood cells in the bone marrow do not mature, and as a result, do not develop into healthy blood cells. Early on, no symptoms typically are seen. Later, symptoms may include fatigue, shortness of breath, bleeding disorders, anemia, or frequent infections. Some types may develop into acute myeloid leukemia.

<span class="mw-page-title-main">Fanconi anemia</span> Medical condition

Fanconi anemia (FA) is a rare, AR, genetic disease resulting in impaired response to DNA damage in the FA/BRCA pathway. Although it is a very rare disorder, study of this and other bone marrow failure syndromes has improved scientific understanding of the mechanisms of normal bone marrow function and development of cancer. Among those affected, the majority develop cancer, most often acute myelogenous leukemia (AML), MDS, and liver tumors. 90% develop aplastic anemia by age 40. About 60–75% have congenital defects, commonly short stature, abnormalities of the skin, arms, head, eyes, kidneys, and ears, and developmental disabilities. Around 75% have some form of endocrine problem, with varying degrees of severity. 60% of FA is FANC-A, 16q24.3, which has later onset bone marrow failure.

<span class="mw-page-title-main">Tumors of the hematopoietic and lymphoid tissues</span> Tumors that affect the blood, bone marrow, lymph, and lymphatic system

Tumors of the hematopoietic and lymphoid tissues or tumours of the haematopoietic and lymphoid tissues are tumors that affect the blood, bone marrow, lymph, and lymphatic system. Because these tissues are all intimately connected through both the circulatory system and the immune system, a disease affecting one will often affect the others as well, making aplasia, myeloproliferation and lymphoproliferation closely related and often overlapping problems. While uncommon in solid tumors, chromosomal translocations are a common cause of these diseases. This commonly leads to a different approach in diagnosis and treatment of hematological malignancies. Hematological malignancies are malignant neoplasms ("cancer"), and they are generally treated by specialists in hematology and/or oncology. In some centers "hematology/oncology" is a single subspecialty of internal medicine while in others they are considered separate divisions. Not all hematological disorders are malignant ("cancerous"); these other blood conditions may also be managed by a hematologist.

Primary myelofibrosis (PMF) is a rare bone marrow blood cancer. It is classified by the World Health Organization (WHO) as a type of myeloproliferative neoplasm, a group of cancers in which there is activation and growth of mutated cells in the bone marrow. This is most often associated with a somatic mutation in the JAK2, CALR, or MPL genes. In PMF, the bony aspects of bone marrow are remodeled in a process called osteosclerosis; in addition, fibroblast secrete collagen and reticulin proteins that are collectively referred to as (fibrosis). These two pathological processes compromise the normal function of bone marrow resulting in decreased production of blood cells such as erythrocytes, granulocytes and megakaryocytes, the latter cells responsible for the production of platelets.

<span class="mw-page-title-main">Acute myeloid leukemia</span> Cancer of the myeloid line of blood cells

Acute myeloid leukemia (AML) is a cancer of the myeloid line of blood cells, characterized by the rapid growth of abnormal cells that build up in the bone marrow and blood and interfere with normal blood cell production. Symptoms may include feeling tired, shortness of breath, easy bruising and bleeding, and increased risk of infection. Occasionally, spread may occur to the brain, skin, or gums. As an acute leukemia, AML progresses rapidly, and is typically fatal within weeks or months if left untreated.

<span class="mw-page-title-main">Chronic myelomonocytic leukemia</span> Medical condition

Chronic myelomonocytic leukemia (CMML) is a type of leukemia, which are cancers of the blood-forming cells of the bone marrow. In adults, blood cells are formed in the bone marrow, by a process that is known as haematopoiesis. In CMML, there are increased numbers of monocytes and immature blood cells (blasts) in the peripheral blood and bone marrow, as well as abnormal looking cells (dysplasia) in at least one type of blood cell.

<span class="mw-page-title-main">CD135</span> Protein-coding gene in the species Homo sapiens

Cluster of differentiation antigen 135 (CD135) also known as fms like tyrosine kinase 3, receptor-type tyrosine-protein kinase FLT3, or fetal liver kinase-2 (Flk2) is a protein that in humans is encoded by the FLT3 gene. FLT3 is a cytokine receptor which belongs to the receptor tyrosine kinase class III. CD135 is the receptor for the cytokine Flt3 ligand (FLT3L).

<span class="mw-page-title-main">ETV6</span> Protein-coding gene in the species Homo sapiens

ETV6 protein is a transcription factor that in humans is encoded by the ETV6 gene. The ETV6 protein regulates the development and growth of diverse cell types, particularly those of hematological tissues. However, its gene, ETV6 frequently suffers various mutations that lead to an array of potentially lethal cancers, i.e., ETV6 is a clinically significant proto-oncogene in that it can fuse with other genes to drive the development and/or progression of certain cancers. However, ETV6 is also an anti-oncogene or tumor suppressor gene in that mutations in it that encode for a truncated and therefore inactive protein are also associated with certain types of cancers.

<span class="mw-page-title-main">RUNX1</span> Protein-coding gene in humans

Runt-related transcription factor 1 (RUNX1) also known as acute myeloid leukemia 1 protein (AML1) or core-binding factor subunit alpha-2 (CBFA2) is a protein that in humans is encoded by the RUNX1 gene.

<span class="mw-page-title-main">HOXA9</span> Protein-coding gene in humans

Homeobox protein Hox-A9 is a protein that in humans is encoded by the HOXA9 gene.

<span class="mw-page-title-main">GATA2</span> Protein-coding gene in the species Homo sapiens

GATA2 or GATA-binding factor 2 is a transcription factor, i.e. a nuclear protein which regulates the expression of genes. It regulates many genes that are critical for the embryonic development, self-renewal, maintenance, and functionality of blood-forming, lympathic system-forming, and other tissue-forming stem cells. GATA2 is encoded by the GATA2 gene, a gene which often suffers germline and somatic mutations which lead to a wide range of familial and sporadic diseases, respectively. The gene and its product are targets for the treatment of these diseases.

<span class="mw-page-title-main">PHF6</span> Protein-coding gene in the species Homo sapiens

PHD finger protein 6 is a protein that in humans is encoded by the PHF6 gene.

<span class="mw-page-title-main">Acute megakaryoblastic leukemia</span> Medical condition

Acute megakaryoblastic leukemia (AMKL) is life-threatening leukemia in which malignant megakaryoblasts proliferate abnormally and injure various tissues. Megakaryoblasts are the most immature precursor cells in a platelet-forming lineage; they mature to promegakaryocytes and, ultimately, megakaryocytes which cells shed membrane-enclosed particles, i.e. platelets, into the circulation. Platelets are critical for the normal clotting of blood. While malignant megakaryoblasts usually are the predominant proliferating and tissue-damaging cells, their similarly malignant descendants, promegakaryocytes and megakaryocytes, are variable contributors to the malignancy.

<span class="mw-page-title-main">Tet methylcytosine dioxygenase 2</span> Human gene

Tet methylcytosine dioxygenase 2 (TET2) is a human gene. It resides at chromosome 4q24, in a region showing recurrent microdeletions and copy-neutral loss of heterozygosity (CN-LOH) in patients with diverse myeloid malignancies.

Graft-versus-tumor effect (GvT) appears after allogeneic hematopoietic stem cell transplantation (HSCT). The graft contains donor T cells that can be beneficial for the recipient by eliminating residual malignant cells. GvT might develop after recognizing tumor-specific or recipient-specific alloantigens. It could lead to remission or immune control of hematologic malignancies. This effect applies in myeloma and lymphoid leukemias, lymphoma, multiple myeloma and possibly breast cancer. It is closely linked with graft-versus-host disease (GvHD), as the underlying principle of alloimmunity is the same. CD4+CD25+ regulatory T cells (Treg) can be used to suppress GvHD without loss of beneficial GvT effect. The biology of GvT response is still not fully understood but it is probable that the reaction with polymorphic minor histocompatibility antigens expressed either specifically on hematopoietic cells or more widely on a number of tissue cells or tumor-associated antigens is involved. This response is mediated largely by cytotoxic T lymphocytes (CTL) but it can be employed by natural killers as separate effectors, particularly in T-cell-depleted HLA-haploidentical HSCT.

<span class="mw-page-title-main">Musashi-2</span> Protein-coding gene in the species Homo sapiens

Musashi-2, also known as Musashi RNA binding protein 2, is a protein that in humans is encoded by the MSI2 gene. Like its homologue musashi-1 (MSI1), it is an RNA-binding protein involved in stemness.

Clonal hypereosinophilia, also termed primary hypereosinophilia or clonal eosinophilia, is a grouping of hematological disorders all of which are characterized by the development and growth of a pre-malignant or malignant population of eosinophils, a type of white blood cell that occupies the bone marrow, blood, and other tissues. This population consists of a clone of eosinophils, i.e. a group of genetically identical eosinophils derived from a sufficiently mutated ancestor cell.

GATA2 deficiency is a grouping of several disorders caused by common defect, namely, familial or sporadic inactivating mutations in one of the two parental GATA2 genes. Being the gene haploinsufficient, mutations that cause a reduction in the cellular levels of the gene's product, GATA2, are autosomal dominant. The GATA2 protein is a transcription factor critical for the embryonic development, maintenance, and functionality of blood-forming, lymphatic-forming, and other tissue-forming stem cells. In consequence of these mutations, cellular levels of GATA2 are deficient and individuals develop over time hematological, immunological, lymphatic, or other presentations that may begin as apparently benign abnormalities but commonly progress to severe organ failure, opportunistic infections, virus infection-induced cancers, the myelodysplastic syndrome, and/or leukemia. GATA2 deficiency is a life-threatening and precancerous condition.

Christopher Hourigan is a physician-scientist known for work on measurable residual disease in acute myeloid leukemia.

References

  1. 1 2 3 4 5 6 7 8 9 10 11 12 Jan, Max; Ebert, Benjamin L.; Jaiswal, Siddhartha (1 January 2017). "Clonal hematopoiesis". Seminars in Hematology. 54 (1): 43–50. doi: 10.1053/j.seminhematol.2016.10.002 . ISSN   1532-8686. PMC   8045769 . PMID   28088988.
  2. Sperling, Adam S.; Gibson, Christopher J.; Ebert, Benjamin L. (2017). "The genetics of myelodysplastic syndrome: from clonal haematopoiesis to secondary leukaemia". Nature Reviews Cancer. 17 (1): 5–19. doi:10.1038/nrc.2016.112. ISSN   1474-1768. PMC   5470392 . PMID   27834397.
  3. 1 2 3 4 5 6 7 8 Steensma, David P.; Bejar, Rafael; Jaiswal, Siddhartha; Lindsley, R. Coleman; Sekeres, Mikkael A.; Hasserjian, Robert P.; Ebert, Benjamin L. (2 July 2015). "Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes". Blood. 126 (1): 9–16. doi:10.1182/blood-2015-03-631747. ISSN   1528-0020. PMC   4624443 . PMID   25931582.
  4. 1 2 3 4 5 6 7 8 9 10 11 12 Genovese, Giulio; Kähler, Anna K.; Handsaker, Robert E.; Lindberg, Johan; Rose, Samuel A.; Bakhoum, Samuel F.; Chambert, Kimberly; Mick, Eran; Neale, Benjamin M.; Fromer, Menachem; Purcell, Shaun M.; Svantesson, Oscar; Landén, Mikael; Höglund, Martin; Lehmann, Sören; Gabriel, Stacey B.; Moran, Jennifer L.; Lander, Eric S.; Sullivan, Patrick F.; Sklar, Pamela; Grönberg, Henrik; Hultman, Christina M.; McCarroll, Steven A. (25 December 2014). "Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence". The New England Journal of Medicine. 371 (26): 2477–2487. doi:10.1056/NEJMoa1409405. ISSN   1533-4406. PMC   4290021 . PMID   25426838.
  5. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 Jaiswal, Siddhartha; Fontanillas, Pierre; Flannick, Jason; Manning, Alisa; Grauman, Peter V.; Mar, Brenton G.; Lindsley, R. Coleman; Mermel, Craig H.; Burtt, Noel; Chavez, Alejandro; Higgins, John M.; Moltchanov, Vladislav; Kuo, Frank C.; Kluk, Michael J.; Henderson, Brian; Kinnunen, Leena; Koistinen, Heikki A.; Ladenvall, Claes; Getz, Gad; Correa, Adolfo; Banahan, Benjamin F.; Gabriel, Stacey; Kathiresan, Sekar; Stringham, Heather M.; McCarthy, Mark I.; Boehnke, Michael; Tuomilehto, Jaakko; Haiman, Christopher; Groop, Leif; Atzmon, Gil; Wilson, James G.; Neuberg, Donna; Altshuler, David; Ebert, Benjamin L. (25 December 2014). "Age-related clonal hematopoiesis associated with adverse outcomes". The New England Journal of Medicine. 371 (26): 2488–2498. doi:10.1056/NEJMoa1408617. ISSN   1533-4406. PMC   4306669 . PMID   25426837.
  6. 1 2 3 4 5 6 7 Xie, Mingchao; Lu, Charles; Wang, Jiayin; McLellan, Michael D.; Johnson, Kimberly J.; Wendl, Michael C.; McMichael, Joshua F.; Schmidt, Heather K.; Yellapantula, Venkata; Miller, Christopher A.; Ozenberger, Bradley A.; Welch, John S.; Link, Daniel C.; Walter, Matthew J.; Mardis, Elaine R.; Dipersio, John F.; Chen, Feng; Wilson, Richard K.; Ley, Timothy J.; Ding, Li (1 December 2014). "Age-related mutations associated with clonal hematopoietic expansion and malignancies". Nature Medicine. 20 (12): 1472–1478. doi:10.1038/nm.3733. ISSN   1546-170X. PMC   4313872 . PMID   25326804.
  7. McKerrell, T; Park, N; Moreno, T; Grove, CS; Ponstingl, H; Stephens, J; Understanding Society Scientific, Group.; Crawley, C; Craig, J; Scott, MA; Hodkinson, C; Baxter, J; Rad, R; Forsyth, DR; Quail, MA; Zeggini, E; Ouwehand, W; Varela, I; Vassiliou, GS (3 March 2015). "Leukemia-associated somatic mutations drive distinct patterns of age-related clonal hemopoiesis". Cell Reports. 10 (8): 1239–45. doi:10.1016/j.celrep.2015.02.005. PMC   4542313 . PMID   25732814.
  8. 1 2 3 4 5 6 Zink, Florian; Stacey, Simon N.; Norddahl, Gudmundur L.; Frigge, Michael L.; Magnusson, Olafur T.; Jonsdottir, Ingileif; Thorgeirsson, Thorgeir E.; Sigurdsson, Asgeir; Gudjonsson, Sigurjon A. (2017-01-01). "Clonal hematopoiesis, with and without candidate driver mutations, is common in the elderly". Blood. 130 (6): blood–2017–02–769869. doi:10.1182/blood-2017-02-769869. ISSN   0006-4971. PMC   5553576 . PMID   28483762.
  9. 1 2 da Silva-Coelho, Pedro; Kroeze, Leonie I.; Yoshida, Kenichi; Koorenhof-Scheele, Theresia N.; Knops, Ruth; van de Locht, Louis T.; de Graaf, Aniek O.; Massop, Marion; Sandmann, Sarah; Dugas, Martin; Stevens-Kroef, Marian J.; Cermak, Jaroslav; Shiraishi, Yuichi; Chiba, Kenichi; Tanaka, Hiroko; Miyano, Satoru; de Witte, Theo; Blijlevens, Nicole M. A.; Muus, Petra; Huls, Gerwin; van der Reijden, Bert A.; Ogawa, Seishi; Jansen, Joop H. (21 April 2017). "Clonal evolution in myelodysplastic syndromes". Nature Communications. 8: 15099. Bibcode:2017NatCo...815099D. doi:10.1038/ncomms15099. ISSN   2041-1723. PMC   5530598 . PMID   28429724.
  10. 1 2 3 Gibson, Christopher J.; Lindsley, R. Coleman; Tchekmedyian, Vatche; Mar, Brenton G.; Shi, Jiantao; Jaiswal, Siddhartha; Bosworth, Alysia; Francisco, Liton; He, Jianbo; Bansal, Anita; Morgan, Elizabeth A.; Lacasce, Ann S.; Freedman, Arnold S.; Fisher, David C.; Jacobsen, Eric; Armand, Philippe; Alyea, Edwin P.; Koreth, John; Ho, Vincent; Soiffer, Robert J.; Antin, Joseph H.; Ritz, Jerome; Nikiforow, Sarah; Forman, Stephen J.; Michor, Franziska; Neuberg, Donna; Bhatia, Ravi; Bhatia, Smita; Ebert, Benjamin L. (9 January 2017). "Clonal Hematopoiesis Associated With Adverse Outcomes After Autologous Stem-Cell Transplantation for Lymphoma". Journal of Clinical Oncology. 35 (14): 1598–1605. doi:10.1200/JCO.2016.71.6712. ISSN   1527-7755. PMC   5455707 . PMID   28068180.
  11. 1 2 3 Jaiswal, Siddhartha; Natarajan, Pradeep; Silver, Alexander J.; Gibson, Christopher J.; Bick, Alexander G.; Shvartz, Eugenia; McConkey, Marie; Gupta, Namrata; Gabriel, Stacey (2017-06-21). "Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease". New England Journal of Medicine. 377 (2): 111–121. doi:10.1056/NEJMoa1701719. ISSN   0028-4793. PMC   6717509 . PMID   28636844.
  12. Park, Soo J.; Bejar, Rafael (2020-03-01). "Clonal hematopoiesis in cancer". Experimental Hematology. 83: 105–112. doi:10.1016/j.exphem.2020.02.001. ISSN   0301-472X. PMC   7103485 . PMID   32044376.
  13. Busque, L.; Mio, R.; Mattioli, J.; Brais, E.; Blais, N.; Lalonde, Y.; Maragh, M.; Gilliland, D. G. (1 July 1996). "Nonrandom X-inactivation patterns in normal females: lyonization ratios vary with age". Blood. 88 (1): 59–65. doi: 10.1182/blood.V88.1.59.59 . ISSN   0006-4971. PMID   8704202.
  14. Champion, K. M.; Gilbert, J. G.; Asimakopoulos, F. A.; Hinshelwood, S.; Green, A. R. (1 June 1997). "Clonal haemopoiesis in normal elderly women: implications for the myeloproliferative disorders and myelodysplastic syndromes". British Journal of Haematology. 97 (4): 920–926. doi:10.1046/j.1365-2141.1997.1933010.x. ISSN   0007-1048. PMID   9217198. S2CID   30067488.
  15. Fialkow, P. J.; Singer, J. W.; Adamson, J. W.; Berkow, R. L.; Friedman, J. M.; Jacobson, R. J.; Moohr, J. W. (5 July 1979). "Acute nonlymphocytic leukemia: expression in cells restricted to granulocytic and monocytic differentiation". The New England Journal of Medicine. 301 (1): 1–5. doi:10.1056/NEJM197907053010101. ISSN   0028-4793. PMID   286882.
  16. Fialkow, P. J.; Singer, J. W.; Adamson, J. W.; Vaidya, K.; Dow, L. W.; Ochs, J.; Moohr, J. W. (1 June 1981). "Acute nonlymphocytic leukemia: heterogeneity of stem cell origin". Blood. 57 (6): 1068–1073. doi: 10.1182/blood.V57.6.1068.bloodjournal5761068 . ISSN   0006-4971. PMID   6939452.
  17. Fialkow, P. J.; Janssen, J. W.; Bartram, C. R. (1 April 1991). "Clonal remissions in acute nonlymphocytic leukemia: evidence for a multistep pathogenesis of the malignancy". Blood. 77 (7): 1415–1417. doi: 10.1182/blood.V77.7.1415.1415 . ISSN   0006-4971. PMID   2009365.
  18. Weissman, Irving (21 September 2005). "Stem cell research: paths to cancer therapies and regenerative medicine". JAMA. 294 (11): 1359–1366. doi:10.1001/jama.294.11.1359. ISSN   1538-3598. PMID   16174694.
  19. Kikushige, Yoshikane; Ishikawa, Fumihiko; Miyamoto, Toshihiro; Shima, Takahiro; Urata, Shingo; Yoshimoto, Goichi; Mori, Yasuo; Iino, Tadafumi; Yamauchi, Takuji; Eto, Tetsuya; Niiro, Hiroaki; Iwasaki, Hiromi; Takenaka, Katsuto; Akashi, Koichi (16 August 2011). "Self-renewing hematopoietic stem cell is the primary target in pathogenesis of human chronic lymphocytic leukemia". Cancer Cell. 20 (2): 246–259. doi: 10.1016/j.ccr.2011.06.029 . ISSN   1878-3686. PMID   21840488.
  20. Damm, Frederik; Mylonas, Elena; Cosson, Adrien; Yoshida, Kenichi; Della Valle, Véronique; Mouly, Enguerran; Diop, M'boyba; Scourzic, Laurianne; Shiraishi, Yuichi; Chiba, Kenichi; Tanaka, Hiroko; Miyano, Satoru; Kikushige, Yoshikane; Davi, Frederick; Lambert, Jérôme; Gautheret, Daniel; Merle-Béral, Hélène; Sutton, Laurent; Dessen, Philippe; Solary, Eric; Akashi, Koichi; Vainchenker, William; Mercher, Thomas; Droin, Nathalie; Ogawa, Seishi; Nguyen-Khac, Florence; Bernard, Olivier A. (1 September 2014). "Acquired initiating mutations in early hematopoietic cells of CLL patients". Cancer Discovery. 4 (9): 1088–1101. doi: 10.1158/2159-8290.CD-14-0104 . ISSN   2159-8290. PMID   24920063.
  21. Chung, Stephen S.; Kim, Eunhee; Park, Jae H.; Chung, Young Rock; Lito, Piro; Teruya-Feldstein, Julie; Hu, Wenhuo; Beguelin, Wendy; Monette, Sebastien; Duy, Cihangir; Rampal, Raajit; Telis, Leon; Patel, Minal; Kim, Min Kyung; Huberman, Kety; Bouvier, Nancy; Berger, Michael F.; Melnick, Ari M.; Rosen, Neal; Tallman, Martin S.; Park, Christopher Y.; Abdel-Wahab, Omar (28 May 2014). "Hematopoietic stem cell origin of BRAFV600E mutations in hairy cell leukemia". Science Translational Medicine. 6 (238): 238ra71. doi:10.1126/scitranslmed.3008004. ISSN   1946-6242. PMC   4501573 . PMID   24871132.
  22. Jan, Max; Snyder, Thomas M.; Corces-Zimmerman, M. Ryan; Vyas, Paresh; Weissman, Irving L.; Quake, Stephen R.; Majeti, Ravindra (29 August 2012). "Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia". Science Translational Medicine. 4 (149): 149ra118. doi:10.1126/scitranslmed.3004315. ISSN   1946-6242. PMC   4045621 . PMID   22932223.
  23. Corces-Zimmerman, M. Ryan; Hong, Wan-Jen; Weissman, Irving L.; Medeiros, Bruno C.; Majeti, Ravindra (18 February 2014). "Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission". Proceedings of the National Academy of Sciences of the United States of America. 111 (7): 2548–2553. Bibcode:2014PNAS..111.2548C. doi: 10.1073/pnas.1324297111 . ISSN   1091-6490. PMC   3932921 . PMID   24550281.
  24. Shlush, Liran I.; Zandi, Sasan; Mitchell, Amanda; Chen, Weihsu Claire; Brandwein, Joseph M.; Gupta, Vikas; Kennedy, James A.; Schimmer, Aaron D.; Schuh, Andre C.; Yee, Karen W.; McLeod, Jessica L.; Doedens, Monica; Medeiros, Jessie J. F.; Marke, Rene; Kim, Hyeoung Joon; Lee, Kwon; McPherson, John D.; Hudson, Thomas J.; Brown, Andrew M. K.; Yousif, Fouad; Trinh, Quang M.; Stein, Lincoln D.; Minden, Mark D.; Wang, Jean C. Y.; Dick, John E. (20 February 2014). "Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia". Nature. 506 (7488): 328–333. Bibcode:2014Natur.506..328S. doi:10.1038/nature13038. ISSN   1476-4687. PMC   4991939 . PMID   24522528.
  25. Busque, Lambert; Patel, Jay P.; Figueroa, Maria E.; Vasanthakumar, Aparna; Provost, Sylvie; Hamilou, Zineb; Mollica, Luigina; Li, Juan; Viale, Agnes; Heguy, Adriana; Hassimi, Maryam; Socci, Nicholas; Bhatt, Parva K.; Gonen, Mithat; Mason, Christopher E.; Melnick, Ari; Godley, Lucy A.; Brennan, Cameron W.; Abdel-Wahab, Omar; Levine, Ross L. (1 November 2012). "Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis". Nature Genetics. 44 (11): 1179–1181. doi:10.1038/ng.2413. ISSN   1546-1718. PMC   3483435 . PMID   23001125.
  26. 1 2 "American FactFinder - Results". factfinder.census.gov. Archived from the original on 14 February 2020. Retrieved 1 May 2017.
  27. 1 2 Young, Andrew L.; Challen, Grant A.; Birmann, Brenda M.; Druley, Todd E. (22 August 2016). "Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults". Nature Communications. 7: 12484. Bibcode:2016NatCo...712484Y. doi:10.1038/ncomms12484. ISSN   2041-1723. PMC   4996934 . PMID   27546487.
  28. Abkowitz, Janis L.; Catlin, Sandra N.; McCallie, Monica T.; Guttorp, Peter (1 October 2002). "Evidence that the number of hematopoietic stem cells per animal is conserved in mammals". Blood. 100 (7): 2665–2667. doi: 10.1182/blood-2002-03-0822 . ISSN   0006-4971. PMID   12239184.
  29. Welch, John S.; Ley, Timothy J.; Link, Daniel C.; Miller, Christopher A.; Larson, David E.; Koboldt, Daniel C.; Wartman, Lukas D.; Lamprecht, Tamara L.; Liu, Fulu; Xia, Jun; Kandoth, Cyriac; Fulton, Robert S.; McLellan, Michael D.; Dooling, David J.; Wallis, John W.; Chen, Ken; Harris, Christopher C.; Schmidt, Heather K.; Kalicki-Veizer, Joelle M.; Lu, Charles; Zhang, Qunyuan; Lin, Ling; O'Laughlin, Michelle D.; McMichael, Joshua F.; Delehaunty, Kim D.; Fulton, Lucinda A.; Magrini, Vincent J.; McGrath, Sean D.; Demeter, Ryan T.; Vickery, Tammi L.; Hundal, Jasreet; Cook, Lisa L.; Swift, Gary W.; Reed, Jerry P.; Alldredge, Patricia A.; Wylie, Todd N.; Walker, Jason R.; Watson, Mark A.; Heath, Sharon E.; Shannon, William D.; Varghese, Nobish; Nagarajan, Rakesh; Payton, Jacqueline E.; Baty, Jack D.; Kulkarni, Shashikant; Klco, Jeffery M.; Tomasson, Michael H.; Westervelt, Peter; Walter, Matthew J.; Graubert, Timothy A.; DiPersio, John F.; Ding, Li; Mardis, Elaine R.; Wilson, Richard K. (20 July 2012). "The origin and evolution of mutations in acute myeloid leukemia". Cell. 150 (2): 264–278. doi:10.1016/j.cell.2012.06.023. ISSN   1097-4172. PMC   3407563 . PMID   22817890.
  30. 1 2 Klein, Allon M.; Simons, Benjamin D. (2011-08-01). "Universal patterns of stem cell fate in cycling adult tissues". Development. 138 (15): 3103–3111. doi: 10.1242/dev.060103 . ISSN   0950-1991. PMID   21750026.
  31. 1 2 Heuser, Michael; Thol, Felicitas; Ganser, Arnold (6 May 2016). "Clonal Hematopoiesis of Indeterminate Potential". Deutsches Ärzteblatt International. 113 (18): 317–322. doi:10.3238/arztebl.2016.0317. ISSN   1866-0452. PMC   4961884 . PMID   27215596.
  32. McKerrell, Thomas; Park, Naomi; Moreno, Thaidy; Grove, Carolyn S.; Ponstingl, Hannes; Stephens, Jonathan; Crawley, Charles; Craig, Jenny; Scott, Mike A.; Hodkinson, Clare; Baxter, Joanna; Rad, Roland; Forsyth, Duncan R.; Quail, Michael A.; Zeggini, Eleftheria; Ouwehand, Willem; Varela, Ignacio; Vassiliou, George S. (3 March 2015). "Leukemia-associated somatic mutations drive distinct patterns of age-related clonal hemopoiesis". Cell Reports. 10 (8): 1239–1245. doi:10.1016/j.celrep.2015.02.005. ISSN   2211-1247. PMC   4542313 . PMID   25732814.
  33. Montalban-Bravo, Guillermo; Garcia-Manero, Guillermo; List, Alan; Kantarjian, Hagop M.; Cortes, Jorge E. (1 June 2016). "Myelodysplastic Syndromes". www.cancernetwork.com.
  34. "Acute Myeloid Leukemia (AML) Number of New Cases and Deaths Per 100,000 People (All Races, Males and Females), Age-Adjusted". NIH Surveillance, Epidemiology, and End Results Program (SEER). Archived from the original on 2017-02-03. Retrieved 2017-05-01.
  35. Yu, B; Roberts, MB; Raffield, LM; Zekavat, SM; Nguyen, NQH; Biggs, ML; Brown, MR; Griffin, G; Desai, P; Correa, A; Morrison, AC; Shah, AM; Niroula, A; Uddin, MM; Honigberg, MC; Ebert, BL; Psaty, BM; Whitsel, EA; Manson, JE; Kooperberg, C; Bick, AG; Ballantyne, CM; Reiner, AP; Natarajan, P; Eaton, CB (6 July 2021). "Supplemental Association of Clonal Hematopoiesis With Incident Heart Failure". Journal of the American College of Cardiology. 78 (1): 42–52. doi: 10.1016/j.jacc.2021.04.085 . PMC   8313294 . PMID   34210413.
  36. Schuermans, A; Vlasschaert, C; Nauffal, V; Cho, SMJ; Uddin, MM; Nakao, T; Niroula, A; Klarqvist, MDR; Weeks, LD; Lin, AE; Saadatagah, S; Lannery, K; Wong, M; Hornsby, W; Lubitz, SA; Ballantyne, C; Jaiswal, S; Libby, P; Ebert, BL; Bick, AG; Ellinor, PT; Natarajan, P; Honigberg, MC (11 November 2023). "Clonal haematopoiesis of indeterminate potential predicts incident cardiac arrhythmias". European Heart Journal. doi:10.1093/eurheartj/ehad670. PMID   37952204.
  37. Fuster, José J.; MacLauchlan, Susan; Zuriaga, María A.; Polackal, Maya N.; Ostriker, Allison C.; Chakraborty, Raja; Wu, Chia-Ling; Sano, Soichi; Muralidharan, Sujatha; Rius, Cristina; Vuong, Jacqueline; Jacob, Sophia; Muralidhar, Varsha; Robertson, Avril A. B.; Cooper, Matthew A.; Andrés, Vicente; Hirschi, Karen K.; Martin, Kathleen A.; Walsh, Kenneth (24 February 2017). "Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice". Science. 355 (6327): 842–847. Bibcode:2017Sci...355..842F. doi:10.1126/science.aag1381. ISSN   1095-9203. PMC   5542057 . PMID   28104796.
  38. Zhu, Yanfang Peipei; Hedrick, Catherine C.; Gaddis, Dalia E. (24 February 2017). "Hematopoietic stem cells gone rogue". Science. 355 (6327): 798–799. Bibcode:2017Sci...355..798Z. doi:10.1126/science.aam7939. ISSN   1095-9203. PMID   28232539. S2CID   206657434.
  39. Tall, Alan R.; Levine, Ross L. (2 March 2017). "Cardiovascular disease: Commonality with cancer". Nature. 543 (7643): 45–47. Bibcode:2017Natur.543...45T. doi:10.1038/nature21505. ISSN   1476-4687. PMID   28225756. S2CID   4412193.